US20150158944A1 - Methods for controlling the galactosylation profile of recombinantly-expressed proteins - Google Patents

Methods for controlling the galactosylation profile of recombinantly-expressed proteins Download PDF

Info

Publication number
US20150158944A1
US20150158944A1 US14/619,799 US201514619799A US2015158944A1 US 20150158944 A1 US20150158944 A1 US 20150158944A1 US 201514619799 A US201514619799 A US 201514619799A US 2015158944 A1 US2015158944 A1 US 2015158944A1
Authority
US
United States
Prior art keywords
galactose
nga2f
sum
media
antibody
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
US14/619,799
Other versions
US9255143B2 (en
Inventor
Cornelia Bengea
Lisa M. Rives
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
AbbVie Inc
Original Assignee
AbbVie Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=46046346&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20150158944(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority to US14/619,799 priority Critical patent/US9255143B2/en
Application filed by AbbVie Inc filed Critical AbbVie Inc
Publication of US20150158944A1 publication Critical patent/US20150158944A1/en
Assigned to ABBVIE, INC. reassignment ABBVIE, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: HOSSLER, PATRICK
Assigned to ABBOTT LABORATORIES reassignment ABBOTT LABORATORIES ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BENGEA, Cornelia, RIVES, LISA M.
Assigned to ABBVIE INC. reassignment ABBVIE INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ABBOTT LABORATORIES
Priority to US15/014,694 priority patent/US9365645B1/en
Publication of US9255143B2 publication Critical patent/US9255143B2/en
Application granted granted Critical
Priority to US15/086,739 priority patent/US9505834B2/en
Priority to US15/345,953 priority patent/US20170051052A1/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/24Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against cytokines, lymphokines or interferons
    • C07K16/241Tumor Necrosis Factors
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/0018Culture media for cell or tissue culture
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12PFERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
    • C12P21/00Preparation of peptides or proteins
    • C12P21/005Glycopeptides, glycoproteins
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/10Immunoglobulins specific features characterized by their source of isolation or production
    • C07K2317/14Specific host cells or culture conditions, e.g. components, pH or temperature
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/21Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/40Immunoglobulins specific features characterized by post-translational modification
    • C07K2317/41Glycosylation, sialylation, or fucosylation
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/05Inorganic components
    • C12N2500/10Metals; Metal chelators
    • C12N2500/20Transition metals
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/30Organic components
    • C12N2500/34Sugars
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2511/00Cells for large scale production

Definitions

  • the present invention relates to methods for modulating the glycosylation profile of recombinantly-expressed proteins.
  • the present invention relates to methods of controlling the galactosylation profile of recombinantly-expressed proteins by supplementing production media with manganese and/or galactose.
  • a particular type of production media e.g., hydrolysate-based media or chemically defined media (“CD” or “CDM”)
  • CD chemically defined media
  • recombinant proteins produced in different CD or hydrolysate-based media can exhibit large differences in their product quality profile. In certain instances, this variability can lead to increases in the fraction of the agalactosyl fucosylated biantennary oligosaccharides NGA2F+NGA2F-GlcNAc and decreases in the fraction of galactose-containing fucosylated biantennary oligosaccharides NA1F+NA2F. Shifts in the glycosylation profile of recombinant proteins of this magnitude are significant as these shifts may render the resulting production lots of the target protein out of compliance with approved process specifications.
  • the present invention relates to methods for modulating the glycosylation profile of recombinantly-expressed proteins.
  • the present invention relates to methods of controlling the galactosylation profile of recombinantly-expressed proteins by supplementing production media with manganese and/or galactose.
  • the production media is a hydrolysate-based media or a CD media.
  • the present invention is directed to methods of controlling the galactosylation profile of recombinantly-expressed antibody.
  • the recombinantly-expressed antibody is an anti-TNF ⁇ antibody.
  • the recombinantly-expressed anti-TNF ⁇ antibody is adalimumab.
  • the present invention is directed to methods of controlling the galactosylation profile of recombinantly-expressed proteins by supplementing a production medium, e.g., a hydrolysate-based or a CD medium, used in the production of recombinantly-expressed proteins with manganese and/or galactose.
  • a production medium e.g., a hydrolysate-based or a CD medium
  • the manganese supplement can take the form of any biologically-acceptable manganese salt, for example, but not limited to, manganese (II) chloride.
  • the galactose supplement can take the form of any biologically-acceptable galactose-containing compound, for example, but not limited to, D-(+)-galactose.
  • the present invention is directed to methods of controlling the galactosylation profile of recombinantly-expressed proteins by supplementing a production medium, e.g., a hydrolysate-based or a CD medium, used in the production of recombinantly-expressed proteins with a sufficient amount of manganese and/or a manganese-containing supplement to achieve the following manganese concentrations in the production media: at least about 0.1, at least about 0.2, at least about 0.5, at least about 1.0, at least about 10, at least about 20, at least about 25, at least about 40, at least about 50, at least about 60, at least about 75, at least about 80, or at least about 100 ⁇ M, wherein that production media is used to dilute a supplement-free cell culture growth media containing no supplement by a ratio of about 1:4 or about 1:5 (supplement-free growth media: supplemented production media).
  • a production medium e.g., a hydrolysate-based or a CD medium
  • the present invention is directed to methods of controlling the galactosylation profile of recombinantly-expressed proteins by supplementing a production medium, e.g., a hydrolysate-based or a CD medium, used in the production of the recombinantly-expressed proteins with sufficient galactose and/or galactose-containing supplement to achieve the following galactose concentrations in the production media: at least about 1, at least about 4, at least about 5, at least about 10, at least about 15, at least about 20, at least about 30, at least about 40, at least about 60, or at least about 100 mM, wherein that production media is used to dilute a supplement-free cell culture growth media containing no supplement by a ratio of about 1:4 or about 1:5 (supplement-free growth media: supplemented production media).
  • a production medium e.g., a hydrolysate-based or a CD medium
  • the present invention is directed to methods of controlling the galactosylation profile of recombinantly-expressed proteins by supplementing a production medium, e.g., a hydrolysate-based or a CD medium, used in the production of recombinantly-expressed proteins with sufficient manganese and/or a manganese-containing supplement and sufficient galactose and/or galactose-containing supplement to achieve at least about the following manganese (Mn) and galactose (Gal) concentrations in the production media presented as Mn ( ⁇ M)/Gal (mM): 0/1, 0/4, 0/5, 0/10, 0/15, 0/20, 0/30, 0/40, 0/60, 0/100, 0.1/0, 0.2/0, 0.5/0, 1.0/0, 10/0, 20/0, 25/0, 40/0, 50/0, 75/0, 80/0, 100/0, 0.2/1
  • the present invention is directed to methods of controlling the galactosylation profile of recombinantly-expressed proteins by supplementing a production medium, e.g., a hydrolysate-based or a CD medium, used in the production of recombinantly-expressed proteins with sufficient manganese and/or a manganese-containing supplement and sufficient galactose and/or galactose-containing supplement to achieve at least about the following manganese (Mn) and galactose (Gal) concentrations in the production media presented as Mn ( ⁇ M)/Gal (mM): 0.2/1, 0.2/4, 0.2/30, 0.5/1, 0.5/4, 0.5/30, 10/10, 10/20, 10/40, 20/10, 20/20, 20/40, 25/15, 40/10, 40/20, 40/40, 40/100, 50/30, 60/20, 60/40, 60/100, 80/20, 80/40, 80/100, 100/20, 100/40, 100/100
  • FIG. 1A depicts the culture growth of adalimumab-producing CHO cell line in CDM GIA-1 in batch shake flasks.
  • FIG. 1B depicts the viability of adalimumab-producing CHO cell line in CDM GIA-1 in batch shake flasks.
  • FIG. 1C depicts the normalized titer of adalimumab-producing CHO cell line in CDM GIA-1 in batch shake flasks.
  • FIG. 2A depicts the culture growth of adalimumab-producing CHO cell line in CDM GIA-1 in fed-batch 3 L bioreactors.
  • FIG. 2B depicts the viability of adalimumab-producing CHO cell line in CDM GIA-1 in fed-batch 3 L bioreactors.
  • FIG. 2C depicts the normalized titer of adalimumab-producing CHO cell line in CDM GIA-1 in fed-batch 3 L bioreactors.
  • FIG. 3A depicts the galactosylation profile (NGA2F+NGA2F-GlcNac) of adalimumab in CHO cell line in CDM GIA-1 in batch shake flasks.
  • FIG. 3B depicts the galactosylation profile (NA1F+NA2F) of adalimumab in CHO cell line in CDM GIA-1 in batch shake flasks.
  • FIG. 4 depicts the percentage galactosylation change of adalimumab in CDM GIA-1 in batch shake flasks relative to control.
  • FIG. 5 summarizes the effect of manganese and/or galactose addition to CDM GIA-1 on galactosylation of adalimumab relative to control in CHO cell line.
  • FIG. 6A depicts the galactosylation profile (NGA2F+NGA2F-GlcNac) of adalimumab in CHO cell line in CDM GIA-1 in fed-batch 3 L bioreactors.
  • FIG. 6B depicts the galactosylation profile (NA1F+NA2F) of adalimumab in CHO cell line in CDM GIA-1 in fed-batch 3 L bioreactors.
  • FIG. 7A depicts the culture growth of CHO cell line in CDM HyClone CDM4CHO in batch shake flasks.
  • FIG. 7B depicts the viability of CHO cell line in CDM HyClone CDM4CHO in batch shake flasks.
  • FIG. 8A depicts the galactosylation profile (NGA2F+NGA2F-GlcNac) of adalimumab in CHO cell line in CDM HyClone CDM4CHO in batch shake flasks.
  • FIG. 8B depicts the galactosylation profile (NA1F+NA2F) of adalimumab in CHO cell line in CDM HyClone CDM4CHO in batch shake flasks.
  • FIG. 9 summarizes the effect of manganese and/or galactose addition to CDM HyClone CDM4CHO on galactosylation of adalimumab relative to control in CHO cell line.
  • FIG. 10A depicts the culture growth of CHO cell line in hydrolysate media in batch shake flasks.
  • FIG. 10B depicts the viability of CHO cell line in hydrolysate media in batch shake flasks.
  • FIG. 11A depicts the galactosylation profile (NGA2F+NGA2F-GlcNac) of adalimumab in CHO cell line in hydrolysate media in batch shake flasks.
  • FIG. 11B depicts the galactosylation profile (NA1F+NA2F) of adalimumab in CHO cell line in hydrolysate media in batch shake flasks.
  • FIG. 12 summarizes the effect of manganese and/or galactose addition to hydrolysate media on galactosylation of adalimumab relative to control in CHO cell line.
  • FIG. 13A depicts the culture growth of adalimumab-producing CHO cell line #2 in CDM GIA-1 in batch shake flasks.
  • FIG. 13B depicts the viability of adalimumab-producing CHO cell line #2 in CDM GIA-1 in batch shake flasks.
  • FIG. 14A depicts the galactosylation profile (NGA2F+NGA2F-GlcNac) of adalimumab in CHO cell line #2 in CDM GIA-1 in batch shake flasks.
  • FIG. 14B depicts the galactosylation profile (NA1F+NA2F) of adalimumab in CHO cell line #2 in CDM GIA-1 in batch shake flasks.
  • FIG. 15 summarizes the effect of manganese and/or galactose addition to CDM GIA-1 on galactosylation of adalimumab relative to control in CHO cell line #2.
  • FIG. 17A depicts the galactosylation profile (NGA2F+NGA2F-GlcNac) of adalimumab in CHO cell line #3 in CDM GIA-1 in fed-batch 3 L bioreactors.
  • FIG. 17B depicts the galactosylation profile (NA1F+NA2F) of adalimumab in CHO cell line #3 in CDM GIA-1 in fed-batch 3 L bioreactors.
  • FIG. 18 summarizes the effect of manganese and/or galactose addition to CDM GIA-1 on galactosylation of adalimumab relative to control in CHO cell line #3.
  • FIG. 19A depicts the culture growth of adalimumab-producing NSO cell line in CDM PFBM-3/PFFM-4 fed-batch shake flasks.
  • FIG. 19B depicts the viability of adalimumab-producing NSO cell line in CDM PFBM-3/PFFM-4 fed-batch shake flasks.
  • FIG. 19C depicts the normalized titer of adalimumab-producing NSO cell line in CDM PFBM-3/PFFM-4 fed-batch shake flasks.
  • FIG. 20A depicts the galactosylation profile (NGA2F+NGA2F-GlcNac) of adalimumab in NSO cell line in CDM PFBM-3/PFFM-4 fed-batch shake flasks.
  • FIG. 20B depicts the galactosylation profile (NA1F+NA2F) of adalimumab in NSO cell line in CDM PFBM-3/PFFM-4 fed-batch shake flasks.
  • FIG. 21 summarizes the effect of manganese and/or galactose addition to CDM PFBM-3/PFFM-4 on galactosylation of adalimumab relative to control in NSO cell line.
  • FIG. 22A depicts the culture growth of CHO cell line producing mAb #1 in CDM GIA-1 in batch shake flasks.
  • FIG. 22B depicts the viability of CHO cell line producing mAb #1 in CDM GIA-1 in batch shake flasks.
  • FIG. 23A depicts the galactosylation profile (NGA2F+NGA2F-GlcNac) of mAb #1 in CDM GIA-1 in batch shake flasks.
  • FIG. 23B depicts the galactosylation profile (NA1F+NA2F) of mAb #1 in CDM GIA-1 in batch shake flasks.
  • FIG. 24 summarizes the effect of manganese and/or galactose addition to CDM GIA-1 on galactosylation of mAb #1 relative to control.
  • FIG. 25A depicts the culture growth of CHO cell line producing mAb #2 in CDM GIA-1 in fed-batch 3 L bioreactors.
  • FIG. 25B depicts the viability of CHO cell line producing mAb #2 in CDM GIA-1 in fed-batch 3 L bioreactors.
  • FIG. 25C depicts the normalized titer of CHO cell line producing mAb #2 in CDM GIA-1 in fed-batch 3 L bioreactors.
  • FIG. 26A depicts the glycosylation profile (NGA2F+NGA2F-GlcNAc) of mAb #2 in CDM GIA-1 in fed-batch 3 L bioreactors.
  • FIG. 26B depicts the glycosylation profile (NA1F+NA2F) of mAb #2 in CDM GIA-1 in fed-batch 3 L bioreactors.
  • FIG. 27 summarizes the effect of manganese and/or galactose addition to CDM GIA-1 on galactosylation of mAb #2 relative to control.
  • the present invention relates to methods modulating the glycosylation profile of recombinantly-expressed proteins.
  • the present invention relates to methods of controlling (e.g., modulating) the galactosylation profile of recombinantly-expressed proteins by supplementing production medium, e.g., a hydrolysate-based or a CD medium, with manganese and/or galactose.
  • production medium e.g., a hydrolysate-based or a CD medium
  • the present invention demonstrates that supplementation of particular ranges of manganese and/or galactose concentrations to chemically defined media can be used to fine-tune the galactosylation profile of monoclonal antibodies produced in CHO and NSO cell lines.
  • a terminal galactose is added to NGA2F by ⁇ -galactosyltransferase enzyme in the presence of manganese chloride, to produce NA1F (in the case of an addition of a single terminal galactose) or NA2F (in the case of an addition of two terminal galactose molecules).
  • This galactosyltransferase-mediated reaction employs UDP-galactose as the sugar substrate and Mn 2+ as a cofactor for galactosyltransferase.
  • the present invention is directed to methods of controlling the galactosylation profile of recombinantly-expressed antibody.
  • the recombinantly-expressed antibody is an anti-TNF ⁇ antibody.
  • the recombinantly-expressed anti-TNF ⁇ antibody is adalimumab.
  • the present invention is directed to methods of controlling the galactosylation profile of recombinantly-expressed proteins by supplementing a production medium, e.g., a hydrolysate-based or a CD medium, used in the production of recombinantly-expressed proteins with manganese and/or galactose.
  • a production medium e.g., a hydrolysate-based or a CD medium
  • the manganese supplement can take the form of any biologically-acceptable manganese salt, for example, but not limited to, manganese (II) chloride.
  • the galactose supplement can take the form of any biologically-acceptable galactose-containing compound, for example, but not limited to, D-(+)-galactose.
  • the present invention is directed to methods of controlling the galactosylation profile of recombinantly-expressed proteins by supplementing a production medium, e.g., a hydrolysate-based or a CD medium, used in the production of recombinantly-expressed proteins with sufficient galactose and/or galactose-containing supplement to achieve at least about the following galactose concentrations in the production media: at least about 1, at least about 4, at least about 5, at least about 10, at least about 15, at least about 20, at least about 30, at least about 40, at least about 60, or at least about 100 mM, wherein that production media is used to dilute a supplement-free cell culture growth media containing no supplement by a ratio of about 1:4 or about 1:5 (supplement-free growth media: supplemented production media).
  • a production medium e.g., a hydrolysate-based or a CD medium
  • the present invention is directed to methods of controlling the galactosylation profile of recombinantly-expressed proteins by supplementing a production medium, e.g., a hydrolysate-based or a CD medium, used in the production of recombinantly-expressed proteins with sufficient manganese and/or a manganese-containing supplement and sufficient galactose and/or galactose-containing supplement to achieve at least about the following manganese (Mn) and galactose (Gal) concentrations in the production media presented as Mn ( ⁇ M)/Gal (mM): 0/1, 0/4, 0/5, 0/10, 0/15, 0/20, 0/30, 0/40, 0/60, 0/100, 0.1/0, 0.2/0, 0.5/0, 1.0/0, 10/0, 20/0, 25/0, 40/0, 50/0, 75/0, 80/0, 100/0, 0.2/1
  • the production medium e.g., a hydrolysate-based or a CD medium
  • the production medium used in the production of a recombinantly-expressed protein
  • addition of manganese and not galactose to production IVGN CDM GIA-1 lowered the NGA2F+NGA2F-GlcNac sum by 6% to 9% and increased the NA1F+NA2F sum by 8% to 9% ( FIGS. 3 , 4 , and 5 ). No further increase in manganese concentration was explored in the experimental design due to the growth inhibition observed at about 100 ⁇ M.
  • the production medium e.g., a hydrolysate-based media or a CD media
  • the production medium used in the production of a recombinantly-expressed protein
  • both manganese and galactose are supplemented with both manganese and galactose.
  • the studies outlined in Example 1 indicate that the addition of combinations of manganese and galactose to production IVGN CDM GIA-1 resulted in a significant decrease in the NGA2F+NGA2F-GlcNac sum of 11% to 26% and a corresponding significant increase in the NA1F+NA2F sum of 13% to 23% as compared to the control condition where no manganese or galactose were added to the production media ( FIGS.
  • the present invention is directed to the supplementation of CD media used in the production of a recombinantly-expressed protein with galactose and/or manganese. That such supplementation is effective across distinct CD media is evidenced by the results outlined in Example 2. Specifically, Example 2 results indicate that the addition of manganese chloride alone within the range of 0 to 40 ⁇ M to production CDM HyClone CDM4CHO decreased the NGA2F+NGA2F-GlcNac sum by a maximum of 5% in a concentration dependent manner ( FIG. 8 ). A comparable maximum increase of 4% in the NA1F+NA2F sum was also achieved.
  • the combined addition of manganese chloride and galactose decreased the NGA2F+NGA2F-GlcNac sum and increased the NA1F+NA2F sum by a comparable percentage as when manganese or galactose were added alone and their individual effects were summed up ( FIG. 9 ).
  • addition of 40 ⁇ M manganese chloride alone reduced the NGA2F+NGA2F-GlcNac sum by 5%
  • addition of 40 mM galactose alone decreased the NGA2F sum by 6%.
  • the combined addition of manganese chloride and galactose at these same concentrations i.e.
  • the present invention is directed to the supplementation of a hydrolysate-based media used in the production of a recombinantly-expressed protein with galactose and/or manganese.
  • a hydrolysate-based media used in the production of a recombinantly-expressed protein with galactose and/or manganese.
  • the addition of manganese chloride alone within the range of 0 to 40 ⁇ M to hydrolysate-based production media decreased the NGA2F+NGA2F-GlcNac sum by approximately 1%, although that change is within the oligosaccharide assay variability ( FIG. 11 ).
  • the combined addition of manganese chloride ranging from 0 to 40 ⁇ M and galactose ranging from 0 to 40 mM to hydrolysate-based media led to an approximate 5% maximum decrease in the NGA2F+NGA2F-GlcNac sum and a corresponding 3% increase in the NA1F+NA2F sum ( FIG. 12 ).
  • the highest percentage decrease of 5% in the NGA2F+NGA2F-GlcNac sum and the corresponding 4% increase in the NA1F+NA2F sum was observed for the culture supplemented with 40 mM galactose and either 20 ⁇ M or 40 ⁇ M manganese chloride.
  • compositions and methods of the present invention also find use across distinct cell lines.
  • the study described in Example 4 illustrates that the supplementation of a CD media, GIA-1, with galactose and/or manganese is effective to modulate galactosylation of adalimumab produced using a CHO cell line distinct from that employed in Examples 1-3.
  • the addition of manganese chloride alone within the range of 0 to 20 ⁇ M to production CDM GIA-1 decreased the NGA2F+NGA2F-GlcNac sum in a concentration dependent manner and increased the NA1F+NA2F sum by approximately the same percentage.
  • Example 5 employs a third adalimumab-producing cell line that is distinct from either of the adalimumab-producing cell lines of Examples 1-4.
  • the addition of manganese chloride alone within the range of 0 to 1 ⁇ M to production CDM GIA-1 decreased the NGA2F+NGA2F-GlcNac sum in a concentration dependent manner and increased the NA1F+NA2F sum by approximately the same percentage.
  • Example 6 which employs a fourth adalimumab-producing cell line that is distinct from the adalimumab-producing cell lines of Examples 1-5, in that it is an NSO cell line.
  • a fourth adalimumab-producing cell line that is distinct from the adalimumab-producing cell lines of Examples 1-5, in that it is an NSO cell line.
  • the addition of manganese chloride alone within the range of 0 to 0.5 ⁇ M to production CDM PFBM-3/PFFM-4 decreased the NGA2F+NGA2F-GlcNac sum in a concentration dependent manner and increased the NA1F+NA2F sum by approximately the same percentage.
  • addition of 0.2 ⁇ M manganese chloride alone reduced the NGA2F+NGA2F-GlcNac sum by 12%
  • addition of 4 mM galactose alone decreased the NGA2F+NGA2F-GlcNac sum by 2%
  • the combined addition of manganese chloride and galactose at these same concentrations led to a 19% reduction in the NGA2F+NGA2F-GlcNac sum, 5% higher than their combined individual contributions.
  • compositions and methods of the present invention also find use in the production of diverse antibodies, as evidenced by the results of Example 7, which employs a CHO cell line that produced an antibody distinct from adalimumab.
  • Example 7 which employs a CHO cell line that produced an antibody distinct from adalimumab.
  • the addition of manganese chloride alone within the range of 0 to 40 ⁇ M to production CDM GIA-1 decreased the NGA2F+NGA2F-GlcNac sum by a maximum of 26% ( FIG. 23 ).
  • a comparable maximum increase of 27% in the NA1F+NA2F sum was also achieved.
  • the addition of 40 ⁇ M manganese chloride alone reduced the NGA2F+NGA2F-GlcNac sum by 20%
  • the addition of 40 mM galactose alone decreased the NGA2F+NGA2F-GlcNac sum by 12%.
  • the combined addition of manganese chloride and galactose at these same concentrations led to a 27% decrease in the NGA2F+NGA2F-GlcNac sum.
  • compositions and methods of the present invention also find use when producing diverse antibodies is further reinforced by the results of Example 8, which employs a CHO cell line producing an antibody distinct from both adalimumab and the antibody of Example 7.
  • Example 8 which employs a CHO cell line producing an antibody distinct from both adalimumab and the antibody of Example 7.
  • the addition of manganese chloride alone in the range of 0 to 75 ⁇ M to production CDM GIA-1 decreased the NGA2F+NGA2F-GlcNac sum by a maximum of 18% ( FIG. 26 ).
  • a comparable maximum increase of 16% in the NA1F+NA2F sum was also achieved.
  • the observed 22% decrease in the NGA2F+NGA2F-GlcNAc sum was 5% more than the sum of the decrease observed with the addition of 25 ⁇ M manganese chloride alone (10%) and 15 mM galactose alone (7%).
  • the additive effect was observed for the condition supplemented with 50 ⁇ M manganese chloride and 30 mM galactose.
  • the observed 28% decrease in the NGA2F+NGA2F-GlcNAc sum was comparable to the sum of the decrease observed with the addition of 50 ⁇ M manganese chloride alone (18%) and 30 mM galactose alone (12%).
  • the expression vector and expression control sequences are chosen to be compatible with the expression host cell used.
  • the antibody light chain gene and the antibody heavy chain gene can be inserted into a separate vector or, more typically, both genes are inserted into the same expression vector.
  • the antibody genes are inserted into an expression vector by standard methods (e.g., ligation of complementary restriction sites on the antibody gene fragment and vector, or blunt end ligation if no restriction sites are present).
  • the expression vector Prior to insertion of the antibody or antibody-related light or heavy chain sequences, the expression vector may already carry antibody constant region sequences.
  • Suitable regulatory sequences for mammalian host cell expression include viral elements that direct high levels of protein expression in mammalian cells, such as promoters and/or enhancers derived from cytomegalovirus (CMV) (such as the CMV promoter/enhancer), Simian Virus 40 (SV40) (such as the SV40 promoter/enhancer), adenovirus, (e.g., the adenovirus major late promoter (AdMLP)) and polyoma.
  • CMV cytomegalovirus
  • SV40 Simian Virus 40
  • AdMLP adenovirus major late promoter
  • the expression vector(s) encoding the heavy and light chains is (are) transfected into a host cell by standard techniques.
  • the various forms of the term “transfection” are intended to encompass a wide variety of techniques commonly used for the introduction of exogenous DNA into a prokaryotic or eukaryotic host cell, e.g., electroporation, calcium-phosphate precipitation, DEAE-dextran transfection and the like.
  • eukaryotic cells such as mammalian host cells
  • expression of antibodies in eukaryotic cells is suitable because such eukaryotic cells, and in particular mammalian cells, are more likely than prokaryotic cells to assemble and secrete a properly folded and immunologically active antibody.
  • Prokaryotic expression of antibody genes has been reported to be ineffective for production of high yields of active antibody (Boss and Wood (1985) Immunology Today 6:12-13, the entire teaching of which is incorporated herein by reference).
  • Suitable host cells for cloning or expressing the DNA in the vectors herein are the prokaryote, yeast, or higher eukaryote cells described above.
  • Suitable prokaryotes for this purpose include eubacteria, such as Gram-negative or Gram-positive organisms, e.g., Enterobacteriaceae such as Escherichia , e.g., E. coli, Enterobacter, Erwinia, Klebsiella, Proteus, Salmonella , e.g., Salmonella typhimurium, Serratia , e.g., Serratia marcescans , and Shigella , as well as Bacilli such as B. subtilis and B.
  • Enterobacteriaceae such as Escherichia , e.g., E. coli, Enterobacter, Erwinia, Klebsiella, Proteus
  • Salmonella e.g., Salmonella typhimurium
  • eukaryotic microbes such as filamentous fungi or yeast are suitable cloning or expression hosts for polypeptide encoding vectors.
  • Saccharomyces cerevisiae or common baker's yeast, is the most commonly used among lower eukaryotic host microorganisms.
  • a number of other genera, species, and strains are commonly available and useful herein, such as Schizosaccharomyces pombe; Kluyveromyces hosts such as, e.g., K. lactis, K. fragilis (ATCC 12,424), K. bulgaricus (ATCC 16,045), K. wickeramii (ATCC 24,178), K.
  • waltii ATCC 56,500
  • K. drosophilarum ATCC 36,906
  • K. thermotolerans K. marxianus
  • yarrowia EP 402,226
  • Pichia pastoris EP 183,070
  • Candida Trichoderma reesia
  • Neurospora crassa Schwanniomyces such as Schwanniomyces occidentalis
  • filamentous fungi such as, e.g., Neurospora, Penicillium, Tolypocladium , and Aspergillus hosts such as A. nidulans and A. niger.
  • Suitable host cells for the expression of glycosylated antibodies are derived from multicellular organisms.
  • invertebrate cells include plant and insect cells.
  • Numerous baculoviral strains and variants and corresponding permissive insect host cells from hosts such as Spodoptera frugiperda (caterpillar), Aedes aegypti (mosquito), Aedes albopictus (mosquito), Drosophila melanogaster (fruitfly), and Bombyx mori have been identified.
  • a variety of viral strains for transfection are publicly available, e.g., the L-1 variant of Autographa californica NPV and the Bm-5 strain of Bombyx mori NPV, and such viruses may be used as the virus herein according to the present invention, particularly for transfection of Spodoptera frugiperda cells.
  • Plant cell cultures of cotton, corn, potato, soybean, petunia, tomato, and tobacco can also be utilized as hosts.
  • Suitable mammalian host cells for expressing the recombinant antibodies of the invention include Chinese Hamster Ovary (CHO cells) (including dhfr-CHO cells, described in Urlaub and Chasin, (1980) PNAS USA 77:4216-4220, used with a DHFR selectable marker, e.g., as described in Kaufman and Sharp (1982) Mol. Biol. 159:601-621, the entire teachings of which are incorporated herein by reference), NS0 myeloma cells, COS cells and SP2 cells.
  • Chinese Hamster Ovary CHO cells
  • dhfr-CHO cells described in Urlaub and Chasin, (1980) PNAS USA 77:4216-4220, used with a DHFR selectable marker, e.g., as described in Kaufman and Sharp (1982) Mol. Biol. 159:601-621, the entire teachings of which are incorporated herein by reference
  • NS0 myeloma cells COS cells and SP2 cells.
  • the antibodies are produced by culturing the host cells for a period of time sufficient to allow for expression of the antibody in the host cells or secretion of the antibody into the culture medium in which the host cells are grown.
  • useful mammalian host cell lines are monkey kidney CV1 line transformed by SV40 (COS-7, ATCC CRL 1651); human embryonic kidney line (293 or 293 cells subcloned for growth in suspension culture, Graham et al., J. Gen Virol. 36:59 (1977)); baby hamster kidney cells (BHK, ATCC CCL 10); Chinese hamster ovary cells/-DHFR (CHO, Urlaub et al., Proc.
  • mice sertoli cells TM4, Mather, Biol. Reprod. 23:243-251 (1980)); monkey kidney cells (CV1 ATCC CCL 70); African green monkey kidney cells (VERO-76, ATCC CRL-1587); human cervical carcinoma cells (HELA, ATCC CCL 2); canine kidney cells (MDCK, ATCC CCL 34); buffalo rat liver cells (BRL 3A, ATCC CRL 1442); human lung cells (W138, ATCC CCL 75); human liver cells (Hep G2, HB 8065); mouse mammary tumor (MMT 060562, ATCC CCL51); TRI cells (Mather et al., Annals N.Y. Acad. Sci. 383:44-68 (1982)); MRC 5 cells; FS4 cells; and a human hepatoma line (Hep G2), the entire teachings of which are incorporated herein by reference.
  • Host cells are transformed with the above-described expression or cloning vectors for antibody production and cultured in conventional nutrient media modified as appropriate for inducing promoters, selecting transformants, or amplifying the genes encoding the desired sequences.
  • the host cells used to produce an antibody may be cultured in a variety of media.
  • Commercially available media such as Ham's F10TM (Sigma), Minimal Essential MediumTM ((MEM), (Sigma), RPMI-1640 (Sigma), and Dulbecco's Modified Eagle's MediumTM ((DMEM), Sigma) are suitable for culturing the host cells.
  • Host cells can also be used to produce portions of intact antibodies, such as Fab fragments or scFv molecules. It is understood that variations on the above procedure are within the scope of the present invention. For example, in certain embodiments it may be desirable to transfect a host cell with DNA encoding either the light chain or the heavy chain (but not both) of an antibody of this invention. Recombinant DNA technology may also be used to remove some or all of the DNA encoding either or both of the light and heavy chains that is not necessary for antigen binding. The molecules expressed from such truncated DNA molecules are also encompassed by the antibodies of the invention.
  • bifunctional antibodies may be produced in which one heavy and one light chain are an antibody of the invention and the other heavy and light chain are specific for an antigen other than the original antigen by crosslinking an antibody of the invention to a second antibody by standard chemical crosslinking methods.
  • a recombinant expression vector encoding both the antibody heavy chain and the antibody light chain is introduced into dhfr-CHO cells by calcium phosphate-mediated transfection.
  • the antibody heavy and light chain genes are each operatively linked to CMV enhancer/AdMLP promoter regulatory elements to drive high levels of transcription of the genes.
  • the recombinant expression vector also carries a DHFR gene, which allows for selection of CHO cells that have been transfected with the vector using methotrexate selection/amplification.
  • the selected transformant host cells are cultured to allow for expression of the antibody heavy and light chains and intact antibody is recovered from the culture medium.
  • Standard molecular biology techniques are used to prepare the recombinant expression vector, transfect the host cells, select for transformants, culture the host cells and recover the antibody from the culture medium.
  • the antibody can be produced intracellularly, in the periplasmic space, or directly secreted into the medium.
  • the particulate debris either host cells or lysed cells (e.g., resulting from homogenization)
  • supernatants from such expression systems can be first concentrated using a commercially available protein concentration filter, e.g., an AmiconTM or Millipore PelliconTM ultrafiltration unit.
  • the first step of a purification process typically involves: lysis of the cell, which can be done by a variety of methods, including mechanical shear, osmotic shock, or enzymatic treatments. Such disruption releases the entire contents of the cell into the homogenate, and in addition produces subcellular fragments that are difficult to remove due to their small size. These are generally removed by differential centrifugation or by filtration.
  • supernatants from such expression systems are generally first concentrated using a commercially available protein concentration filter, e.g., an AmiconTM or Millipore PelliconTM ultrafiltration unit.
  • a commercially available protein concentration filter e.g., an AmiconTM or Millipore PelliconTM ultrafiltration unit.
  • the recombinant host cells can also be separated from the cell culture medium, e.g., by tangential flow filtration.
  • Antibodies can be further recovered from the culture medium using the antibody purification methods of the invention.
  • the cell line utilized for both studies was generated from the adalimumab-producing CHO cell utilized in Example 3 by adapting it to chemically defined GIA-1 media for 7 (2 to 3 day each) passages in a combination of 250 mL and 500 mL Corning vented non-baffled shake flasks before freezing.
  • samples were collected daily and measured for cell density and viability using a Cedex cell counter.
  • Retention samples for titer analysis via Poros A method were collected by centrifugation at 12,000 RPM for 5 min when the culture viability began declining.
  • the cultures were harvested by collecting 125 mL aliquots and centrifuging at 3,000 RPM for 30 min when culture viability was near or below 50%. All supernatants were stored at ⁇ 80° C. until analysis.
  • the harvest samples were Protein A purified and prepared for the oligosaccharide assay using the following procedures.
  • they are released from the protein by enzymatic digestion with N-glycanase. Once the glycans are released, the free reducing end of each glycan is labeled by reductive amination with a fluorescent tag, 2-aminobenzamide (2-AB).
  • the resulting labeled glycans are separated by normal-phase HPLC (NP-HPLC) in acetonitrile: 50 mM ammonium formate, pH 4.4, and detected by a fluorescence detector.
  • Quantitation is based on the relative area percent of detected sugars.
  • the relative area percentages of the agalactosyl fucosylated biantennary oligosaccharides denoted as NGA2F+NGA2F-GlcNAc sum, and the galactose-containing fucosylated biantennary oligosaccharides NA1F+NA2F sum are reported and discussed.
  • manganese chloride was supplemented at the following concentrations in production media: 0, 40, 60, 80, and 100 ⁇ M.
  • Galactose was supplemented at the following levels in production media: 0, 10, 20, 40, and 100 mM.
  • Individual and combined additions of manganese chloride and galactose were studied using a comprehensive design divided into 3 sets of experiments. Each experiment had a control culture for direct comparison of culture growth, productivity, and product quality. Production media used for the control cultures was not supplemented with manganese chloride or galactose. Culture growth, productivity, and product quality data for control cultures is the average of the 3 experiments.
  • manganese chloride and galactose were supplemented to both production and feed media in the following combinations: 40 ⁇ M manganese chloride and 20 mM galactose; 40 ⁇ M manganese chloride and 40 mM galactose (Table 2). Basal and feed media for the control cultures were not supplemented with manganese chloride or galactose.
  • manganese chloride was supplemented at the following concentrations in production media: 0, 10, 20, and 40 ⁇ M.
  • Galactose was supplemented at the following levels in production media: 0, 10, 20, and 100 mM.
  • Production media for the control cultures was not supplemented with manganese chloride or galactose.
  • Growth and production media for the adalimumab-producing CHO cell line were prepared using yeast and soy hydrolysates according to a proprietary formulation. Production media was supplemented with Manganese (II) Chloride (Sigma M1787-100 mL; 1.0 M ⁇ 0.1 M) and D(+)Galactose (Sigma G5388-1 kg) according to the experimental design described in Table 4. All media were filtered through Corning 0.5 L or 1 L filter systems (0.22 ⁇ m PES) and stored at 4° C. until use.
  • Manganese (II) Chloride Sigma M1787-100 mL; 1.0 M ⁇ 0.1 M
  • D(+)Galactose Sigma G5388-1 kg
  • manganese chloride was supplemented at the following concentrations in production media: 0, 10, 20, and 40 ⁇ M.
  • Galactose was supplemented at the following levels in production media: 0, 10, 20, and 40 mM.
  • Production media for the control cultures was not supplemented with manganese chloride or galactose.
  • the oligosaccharide profile changes achieved with the addition of galactose alone are comparable to the changes recorded when combinations of galactose and manganese chloride were added to the hydrolysate-based media.
  • the combined addition of manganese chloride ranging from 0 to 40 ⁇ M and galactose ranging from 0 to 40 mM to hydrolysate-based media led to an approximate 5% maximum decrease in the NGA2F+NGA2F-GlcNac sum and a corresponding 3% increase in the NA1F+NA2F sum ( FIG. 12 ).
  • Growth and production media for the adalimumab-producing CHO cell line #2 were prepared using a proprietary Life Technologies Gibco chemically defined media, GIA-1. Production media only was supplemented with Manganese (II) Chloride (Sigma M1787-100 mL; 1.0 M ⁇ 0.1 M) and D(+)Galactose (Sigma G5388-1 kg) according to the experimental design described in Table 5. All media were filtered through Corning 0.5 L or 1 L filter systems (0.22 ⁇ m PES) and stored at 4° C. until use.
  • Manganese (II) Chloride Sigma M1787-100 mL; 1.0 M ⁇ 0.1 M
  • D(+)Galactose Sigma G5388-1 kg
  • manganese chloride was supplemented at the following concentrations in production media: 0, 10, 20, and 40 ⁇ M.
  • Galactose was supplemented at the following levels in production media: 0, 10, and 20 mM.
  • Production media for the control cultures was not supplemented with manganese chloride or galactose. This study was run in 2 blocks.
  • Growth and production media for the adalimumab-producing CHO cell line #3 were prepared using the proprietary Life Technologies Gibco chemically defined media, GIA-1. Basal production and feed media were supplemented with Manganese (II) Chloride (Sigma M3634-100 g) and D(+)Galactose (Sigma G5388-1 kg) according to the experimental design described in Table 6.
  • Manganese (II) Chloride Sigma M3634-100 g
  • D(+)Galactose Sigma G5388-1 kg
  • the chemically-defined feed from Life Technologies Gibco JCL-5 was added as follows: 4% (v/v)—day 2, 6%—day 4, 8%—day 6, 10%—day 8, and 10%—day 10. Additional 400 g/L glucose was added to the reactor cultures as needed to ensure glucose levels did not deplete. Bioreactors were harvested at a viability of approximately 50% or on production day 17, whichever condition occurred first.
  • manganese chloride was supplemented at the following concentrations in both production and feed media: 0, 0.1, 0.2, 0.5, and 1.0 ⁇ M.
  • Galactose was supplemented at 0 and 30 mM concentrations in both production and feed media.
  • a combined manganese chloride and galactose supplementation strategy was utilized for the production basal and feed media at either 0.2 or 0.5 ⁇ M manganese chloride plus 30 mM galactose. Basal and feed media for the control cultures were not supplemented with manganese chloride or galactose.
  • a maximum decrease of 26% in the NGA2F+NGA2F-GlcNac and a corresponding increase of 28% in the NA1F+NA2F oligosaccharides were observed with the addition of 1 ⁇ M manganese chloride ( FIG. 17 ).
  • Samples were collected every 2 days and measured for cell density and viability using a Cedex cell counter. Retention samples for titer analysis via Poros A method were collected daily beginning on Day 8 by centrifugation at 2,000 g for 10 min and then filtered through 0.2 um PVDF syringe filter. The cultures were harvested on production day 10. The entire culture was collected, chilled on ice to approximately 0° C. for 1.5 hours, the cells and debris flocculated at pH 5.0 by the addition of 1M citric acid and held for 15 minutes, and centrifuged at 4000 ⁇ g for 15 min at 5° C.
  • the supernatant was passed through 0.20 um Millipore Stericup PES filters, and, immediately post filtration, the acidified clarified cell-free harvest was neutralized with 2M Tris to pH 7.1 ⁇ 0.2.
  • the cell free harvest was transferred to PETG bottles and stored at ⁇ 80° C. until analysis.
  • manganese chloride was supplemented at the following concentrations in both production and feed media: 0, 0.2, and 0.5 ⁇ M.
  • Galactose was supplemented at the following levels in both production and feed media: 0, 1, 4, 5, and 10 mM.
  • Manganese chloride and galactose were added in a full factorial, two level DOE design for the 0, 1, and 4 mM galactose conditions and all concentrations of manganese chloride. Individual and combined additions of manganese chloride and galactose were studied using a comprehensive design divided into 2 sets of experiments. Each experiment had a control culture for direct comparison of culture growth, productivity, and product quality. Production medium for control cultures was not supplemented with manganese chloride or galactose.
  • FIGS. 19A , 19 B Culture growth and viability profiles in production media supplemented with galactose in the 0 to 5 mM concentration range and/or manganese chloride up to 0.5 ⁇ M concentration were comparable to the control condition without manganese or galactose added.
  • Harvest titer for most experimental conditions was comparable to the harvest titer for the control condition except for the titer of the culture supplemented with 10 mM galactose, which was 60% lower ( FIG. 19C ).
  • Growth and production media for the CHO cell line producing mAb #1 were prepared using a proprietary Life Technologies Gibco chemically defined media, GIA-1. Production media only was supplemented with Manganese (II) Chloride (Sigma M1787-100 mL; 1.0 M ⁇ 0.1 M) and D(+)Galactose (Sigma G5388-1 kg) according to the experimental design described in Table 8. All media were filtered through Corning 0.5 L or 1 L filter systems (0.22 ⁇ m PES) and stored at 4° C. until use.
  • Manganese (II) Chloride Sigma M1787-100 mL; 1.0 M ⁇ 0.1 M
  • D(+)Galactose Sigma G5388-1 kg
  • manganese chloride was supplemented at the following levels in production media: 0, 10, 20, and 40 ⁇ M.
  • Galactose was supplemented at the following levels in production media: 0, 10, 20, and 100 mM.
  • Production media for the control cultures was not supplemented with manganese chloride or galactose.
  • the combined addition of galactose and manganese chloride to production CDM GIA-1 resulted in a greater percent reduction in the NGA2F+NGA2F-GlcNac sum and, correspondingly, a greater percent increase in the NA1F+NA2F sum as compared to the addition of either component alone ( FIG. 24 ).
  • the addition of 40 ⁇ M manganese chloride alone reduced the NGA2F+NGA2F-GlcNac sum by 20%
  • the addition of 40 mM galactose alone decreased the NGA2F+NGA2F-GlcNac sum by 12%.
  • the combined addition of manganese chloride and galactose at these same concentrations i.e.
  • manganese chloride was supplemented at the following concentrations in both production and feed media: 0, 25, 50, and 75 ⁇ M.
  • Galactose was supplemented at 0, 15, 30, and 60 mM concentrations in both production and feed media.
  • a combined manganese chloride and galactose supplementation strategy was utilized for the production basal and feed media at 25 ⁇ M manganese chloride+15 mM galactose, and 50 ⁇ M manganese chloride+30 mM galactose. Basal and feed media for the control cultures were not supplemented with manganese chloride or galactose.
  • the additive effect was observed for the condition supplemented with 50 ⁇ M manganese chloride and 30 mM galactose.
  • the observed 28% decrease in the NGA2F+NGA2F-GlcNAc sum was comparable to the sum of the decrease observed with the addition of 50 ⁇ M manganese chloride alone (18%) and 30 mM galactose alone (12%).
  • a maximum decrease of 28% in the NGA2F+NGA2F-GlcNac and a corresponding 25% maximum increase in the NA1F+NA2F sum compared to the control condition was observed with the combined addition of 50 ⁇ M manganese chloride and 30 mM galactose to chemically defined GIA-1 media.

Abstract

The present invention relates to methods for modulating the glycosylation profile of recombinantly-expressed proteins. In particular, the present invention relates to methods of controlling the galactosylation profile of recombinantly-expressed proteins by supplementing production medium, e.g., a hydrolysate-based or a chemically defined medium, with manganese and/or D-galactose.

Description

    RELATED APPLICATIONS
  • This application is a continuation application of U.S. patent application Ser. No. 14/493,068, filed on Sep. 22, 2014; which is a divisional application of U.S. patent application Ser. No. 13/457,020, filed on Apr. 26, 2012; which claims the benefit of U.S. Provisional Application Ser. No. 61/479,727, filed on Apr. 27, 2011. The entire contents of each of the foregoing applications are incorporated herein by reference.
  • 1. INTRODUCTION
  • The present invention relates to methods for modulating the glycosylation profile of recombinantly-expressed proteins. In particular, the present invention relates to methods of controlling the galactosylation profile of recombinantly-expressed proteins by supplementing production media with manganese and/or galactose.
  • 2. BACKGROUND OF THE INVENTION
  • Utilization of a particular type of production media, e.g., hydrolysate-based media or chemically defined media (“CD” or “CDM”), for CHO cell cultures producing recombinant proteins can enhance cell growth and target protein production. However, recombinant proteins produced in different CD or hydrolysate-based media can exhibit large differences in their product quality profile. In certain instances, this variability can lead to increases in the fraction of the agalactosyl fucosylated biantennary oligosaccharides NGA2F+NGA2F-GlcNAc and decreases in the fraction of galactose-containing fucosylated biantennary oligosaccharides NA1F+NA2F. Shifts in the glycosylation profile of recombinant proteins of this magnitude are significant as these shifts may render the resulting production lots of the target protein out of compliance with approved process specifications.
  • 3. SUMMARY OF THE INVENTION
  • The present invention relates to methods for modulating the glycosylation profile of recombinantly-expressed proteins. In particular, the present invention relates to methods of controlling the galactosylation profile of recombinantly-expressed proteins by supplementing production media with manganese and/or galactose. In certain embodiments the production media is a hydrolysate-based media or a CD media.
  • In certain embodiments, the present invention is directed to methods of controlling the galactosylation profile of recombinantly-expressed antibody. In certain embodiments, the recombinantly-expressed antibody is an anti-TNFα antibody. In certain embodiments, the recombinantly-expressed anti-TNFα antibody is adalimumab.
  • In certain embodiments, the present invention is directed to methods of controlling the galactosylation profile of recombinantly-expressed proteins by supplementing a production medium, e.g., a hydrolysate-based or a CD medium, used in the production of recombinantly-expressed proteins with manganese and/or galactose. In certain embodiments, the manganese supplement can take the form of any biologically-acceptable manganese salt, for example, but not limited to, manganese (II) chloride. In certain embodiments, the galactose supplement can take the form of any biologically-acceptable galactose-containing compound, for example, but not limited to, D-(+)-galactose.
  • In certain embodiments, the present invention is directed to methods of controlling the galactosylation profile of recombinantly-expressed proteins by supplementing a production medium, e.g., a hydrolysate-based or a CD medium, used in the production of recombinantly-expressed proteins with a sufficient amount of manganese and/or a manganese-containing supplement to achieve the following manganese concentrations in the production media: at least about 0.1, at least about 0.2, at least about 0.5, at least about 1.0, at least about 10, at least about 20, at least about 25, at least about 40, at least about 50, at least about 60, at least about 75, at least about 80, or at least about 100 μM, wherein that production media is used to dilute a supplement-free cell culture growth media containing no supplement by a ratio of about 1:4 or about 1:5 (supplement-free growth media: supplemented production media). In certain embodiments, the present invention is directed to methods of controlling the galactosylation profile of recombinantly-expressed proteins by supplementing a production medium, e.g., a hydrolysate-based or a CD medium, used in the production of the recombinantly-expressed proteins with sufficient galactose and/or galactose-containing supplement to achieve the following galactose concentrations in the production media: at least about 1, at least about 4, at least about 5, at least about 10, at least about 15, at least about 20, at least about 30, at least about 40, at least about 60, or at least about 100 mM, wherein that production media is used to dilute a supplement-free cell culture growth media containing no supplement by a ratio of about 1:4 or about 1:5 (supplement-free growth media: supplemented production media).
  • In certain embodiments, the present invention is directed to methods of controlling the galactosylation profile of recombinantly-expressed proteins by supplementing a production medium, e.g., a hydrolysate-based or a CD medium, used in the production of recombinantly-expressed proteins with sufficient manganese and/or a manganese-containing supplement and sufficient galactose and/or galactose-containing supplement to achieve at least about the following manganese (Mn) and galactose (Gal) concentrations in the production media presented as Mn (μM)/Gal (mM): 0/1, 0/4, 0/5, 0/10, 0/15, 0/20, 0/30, 0/40, 0/60, 0/100, 0.1/0, 0.2/0, 0.5/0, 1.0/0, 10/0, 20/0, 25/0, 40/0, 50/0, 75/0, 80/0, 100/0, 0.2/1, 0.2/4, 0.2/30, 0.5/1, 0.5/4, 0.5/30, 10/10, 10/20, 10/40, 20/10, 20/20, 20/40, 25/15, 40/10, 40/20, 40/40, 40/100, 50/30, 60/20, 60/40, 60/100, 80/20, 80/40, 80/100, 100/20, 100/40, 100/100, wherein that production media is used to dilute a supplement-free cell culture growth media containing no supplement by a ratio of about 1:4 or about 1:5 (supplement-free growth media: supplemented production media).
  • In certain embodiments, the present invention is directed to methods of controlling the galactosylation profile of recombinantly-expressed proteins by supplementing a production medium, e.g., a hydrolysate-based or a CD medium, used in the production of recombinantly-expressed proteins with sufficient manganese and/or a manganese-containing supplement and sufficient galactose and/or galactose-containing supplement to achieve at least about the following manganese (Mn) and galactose (Gal) concentrations in the production media presented as Mn (μM)/Gal (mM): 0.2/1, 0.2/4, 0.2/30, 0.5/1, 0.5/4, 0.5/30, 10/10, 10/20, 10/40, 20/10, 20/20, 20/40, 25/15, 40/10, 40/20, 40/40, 40/100, 50/30, 60/20, 60/40, 60/100, 80/20, 80/40, 80/100, 100/20, 100/40, 100/100, wherein that production media is used to dilute a supplement-free cell culture growth media containing no supplement by a ratio of about 1:4 or about 1:5 (supplement-free growth media: supplemented production media).
  • 4. BRIEF DESCRIPTION OF THE FIGURES
  • FIG. 1A depicts the culture growth of adalimumab-producing CHO cell line in CDM GIA-1 in batch shake flasks. FIG. 1B depicts the viability of adalimumab-producing CHO cell line in CDM GIA-1 in batch shake flasks. FIG. 1C depicts the normalized titer of adalimumab-producing CHO cell line in CDM GIA-1 in batch shake flasks.
  • FIG. 2A depicts the culture growth of adalimumab-producing CHO cell line in CDM GIA-1 in fed-batch 3 L bioreactors. FIG. 2B depicts the viability of adalimumab-producing CHO cell line in CDM GIA-1 in fed-batch 3 L bioreactors. FIG. 2C depicts the normalized titer of adalimumab-producing CHO cell line in CDM GIA-1 in fed-batch 3 L bioreactors.
  • FIG. 3A depicts the galactosylation profile (NGA2F+NGA2F-GlcNac) of adalimumab in CHO cell line in CDM GIA-1 in batch shake flasks. FIG. 3B depicts the galactosylation profile (NA1F+NA2F) of adalimumab in CHO cell line in CDM GIA-1 in batch shake flasks.
  • FIG. 4 depicts the percentage galactosylation change of adalimumab in CDM GIA-1 in batch shake flasks relative to control.
  • FIG. 5 summarizes the effect of manganese and/or galactose addition to CDM GIA-1 on galactosylation of adalimumab relative to control in CHO cell line.
  • FIG. 6A depicts the galactosylation profile (NGA2F+NGA2F-GlcNac) of adalimumab in CHO cell line in CDM GIA-1 in fed-batch 3 L bioreactors. FIG. 6B depicts the galactosylation profile (NA1F+NA2F) of adalimumab in CHO cell line in CDM GIA-1 in fed-batch 3 L bioreactors.
  • FIG. 7A depicts the culture growth of CHO cell line in CDM HyClone CDM4CHO in batch shake flasks. FIG. 7B depicts the viability of CHO cell line in CDM HyClone CDM4CHO in batch shake flasks.
  • FIG. 8A depicts the galactosylation profile (NGA2F+NGA2F-GlcNac) of adalimumab in CHO cell line in CDM HyClone CDM4CHO in batch shake flasks. FIG. 8B depicts the galactosylation profile (NA1F+NA2F) of adalimumab in CHO cell line in CDM HyClone CDM4CHO in batch shake flasks.
  • FIG. 9 summarizes the effect of manganese and/or galactose addition to CDM HyClone CDM4CHO on galactosylation of adalimumab relative to control in CHO cell line.
  • FIG. 10A depicts the culture growth of CHO cell line in hydrolysate media in batch shake flasks. FIG. 10B depicts the viability of CHO cell line in hydrolysate media in batch shake flasks.
  • FIG. 11A depicts the galactosylation profile (NGA2F+NGA2F-GlcNac) of adalimumab in CHO cell line in hydrolysate media in batch shake flasks. FIG. 11B depicts the galactosylation profile (NA1F+NA2F) of adalimumab in CHO cell line in hydrolysate media in batch shake flasks.
  • FIG. 12 summarizes the effect of manganese and/or galactose addition to hydrolysate media on galactosylation of adalimumab relative to control in CHO cell line.
  • FIG. 13A depicts the culture growth of adalimumab-producing CHO cell line #2 in CDM GIA-1 in batch shake flasks. FIG. 13B depicts the viability of adalimumab-producing CHO cell line #2 in CDM GIA-1 in batch shake flasks.
  • FIG. 14A depicts the galactosylation profile (NGA2F+NGA2F-GlcNac) of adalimumab in CHO cell line #2 in CDM GIA-1 in batch shake flasks. FIG. 14B depicts the galactosylation profile (NA1F+NA2F) of adalimumab in CHO cell line #2 in CDM GIA-1 in batch shake flasks.
  • FIG. 15 summarizes the effect of manganese and/or galactose addition to CDM GIA-1 on galactosylation of adalimumab relative to control in CHO cell line #2.
  • FIG. 16A depicts the culture growth of adalimumab-producing CHO cell line #3 in CDM GIA-1 in fed-batch 3 L bioreactors. FIG. 16B depicts the viability of adalimumab-producing CHO cell line #3 in CDM GIA-1 in fed-batch 3 L bioreactors. FIG. 16C depicts the normalized titer of adalimumab-producing CHO cell line #3 in CDM GIA-1 in fed-batch 3 L bioreactors.
  • FIG. 17A depicts the galactosylation profile (NGA2F+NGA2F-GlcNac) of adalimumab in CHO cell line #3 in CDM GIA-1 in fed-batch 3 L bioreactors. FIG. 17B depicts the galactosylation profile (NA1F+NA2F) of adalimumab in CHO cell line #3 in CDM GIA-1 in fed-batch 3 L bioreactors.
  • FIG. 18 summarizes the effect of manganese and/or galactose addition to CDM GIA-1 on galactosylation of adalimumab relative to control in CHO cell line #3.
  • FIG. 19A depicts the culture growth of adalimumab-producing NSO cell line in CDM PFBM-3/PFFM-4 fed-batch shake flasks. FIG. 19B depicts the viability of adalimumab-producing NSO cell line in CDM PFBM-3/PFFM-4 fed-batch shake flasks. FIG. 19C depicts the normalized titer of adalimumab-producing NSO cell line in CDM PFBM-3/PFFM-4 fed-batch shake flasks.
  • FIG. 20A depicts the galactosylation profile (NGA2F+NGA2F-GlcNac) of adalimumab in NSO cell line in CDM PFBM-3/PFFM-4 fed-batch shake flasks. FIG. 20B depicts the galactosylation profile (NA1F+NA2F) of adalimumab in NSO cell line in CDM PFBM-3/PFFM-4 fed-batch shake flasks.
  • FIG. 21 summarizes the effect of manganese and/or galactose addition to CDM PFBM-3/PFFM-4 on galactosylation of adalimumab relative to control in NSO cell line.
  • FIG. 22A depicts the culture growth of CHO cell line producing mAb #1 in CDM GIA-1 in batch shake flasks. FIG. 22B depicts the viability of CHO cell line producing mAb #1 in CDM GIA-1 in batch shake flasks.
  • FIG. 23A depicts the galactosylation profile (NGA2F+NGA2F-GlcNac) of mAb #1 in CDM GIA-1 in batch shake flasks. FIG. 23B depicts the galactosylation profile (NA1F+NA2F) of mAb #1 in CDM GIA-1 in batch shake flasks.
  • FIG. 24 summarizes the effect of manganese and/or galactose addition to CDM GIA-1 on galactosylation of mAb #1 relative to control.
  • FIG. 25A depicts the culture growth of CHO cell line producing mAb #2 in CDM GIA-1 in fed-batch 3 L bioreactors. FIG. 25B depicts the viability of CHO cell line producing mAb #2 in CDM GIA-1 in fed-batch 3 L bioreactors. FIG. 25C depicts the normalized titer of CHO cell line producing mAb #2 in CDM GIA-1 in fed-batch 3 L bioreactors.
  • FIG. 26A depicts the glycosylation profile (NGA2F+NGA2F-GlcNAc) of mAb #2 in CDM GIA-1 in fed-batch 3 L bioreactors. FIG. 26B depicts the glycosylation profile (NA1F+NA2F) of mAb #2 in CDM GIA-1 in fed-batch 3 L bioreactors.
  • FIG. 27 summarizes the effect of manganese and/or galactose addition to CDM GIA-1 on galactosylation of mAb #2 relative to control.
  • 5. DETAILED DESCRIPTION OF THE INVENTION
  • The present invention relates to methods modulating the glycosylation profile of recombinantly-expressed proteins. In particular, the present invention relates to methods of controlling (e.g., modulating) the galactosylation profile of recombinantly-expressed proteins by supplementing production medium, e.g., a hydrolysate-based or a CD medium, with manganese and/or galactose. For example, but not by way of limitation, the present invention demonstrates that supplementation of particular ranges of manganese and/or galactose concentrations to chemically defined media can be used to fine-tune the galactosylation profile of monoclonal antibodies produced in CHO and NSO cell lines. Similarly, supplementation of galactose alone to hydrolysate-based media is effective to modulate the galactosylation profile of the monoclonal antibody adalimumab produced in a CHO cell line in a concentration dependent manner. In view of such findings, the methods disclosed herein can be used to modulate the galactose content of recombinant proteins by controlling the amounts of manganese and/or galactose present in cell culture media. The studies described herein have also established that the changes in the galactosylation profiles obtained via implementation of the methods of the present invention are not only scale (1.5 L vs. 200 mL) and process independent (fed-batch in controlled bioreactor environment vs. batch in shake flasks), but also that no significant impact on culture growth and productivity is observed for most conditions studied.
  • A terminal galactose is added to NGA2F by β-galactosyltransferase enzyme in the presence of manganese chloride, to produce NA1F (in the case of an addition of a single terminal galactose) or NA2F (in the case of an addition of two terminal galactose molecules). This galactosyltransferase-mediated reaction employs UDP-galactose as the sugar substrate and Mn2+ as a cofactor for galactosyltransferase. Thus, without being bound by theory, it is believed that a change in protein homogeneity taking the form of an increase in the fraction of N-linked oligosaccharide NGA2F and a decrease in the fraction of NA1F+NA2F N-linked oligosaccharides could be caused by either an insufficient amount of the substrate (UDP-galactose), the cofactor for galactosyltransferase (Mn2+), or both.
  • In certain embodiments, the present invention is directed to methods of controlling the galactosylation profile of recombinantly-expressed antibody. In certain embodiments, the recombinantly-expressed antibody is an anti-TNFα antibody. In certain embodiments, the recombinantly-expressed anti-TNFα antibody is adalimumab.
  • In certain embodiments, the present invention is directed to methods of controlling the galactosylation profile of recombinantly-expressed proteins by supplementing a production medium, e.g., a hydrolysate-based or a CD medium, used in the production of recombinantly-expressed proteins with manganese and/or galactose. In certain embodiments, the manganese supplement can take the form of any biologically-acceptable manganese salt, for example, but not limited to, manganese (II) chloride. In certain embodiments, the galactose supplement can take the form of any biologically-acceptable galactose-containing compound, for example, but not limited to, D-(+)-galactose.
  • In certain embodiments, the present invention is directed to methods of controlling the galactosylation profile of recombinantly-expressed proteins by supplementing a production medium, e.g., a hydrolysate-based or a CD medium, used in the production of recombinantly-expressed proteins with a sufficient amount of manganese and/or a manganese-containing supplement to achieve at least about the following manganese concentrations in the production media: at least about 0.1, at least about 0.2, at least about 0.5, at least about 1.0, at least about 10, at least about 20, at least about 25, at least about 40, at least about 50, at least about 60, at least about 75, at least about 80, or at least about 100 μM, wherein that production media is used to dilute a supplement-free cell culture growth media containing no supplement by a ratio of about 1:4 or about 1:5 (supplement-free growth media: supplemented production media). In certain embodiments, the present invention is directed to methods of controlling the galactosylation profile of recombinantly-expressed proteins by supplementing a production medium, e.g., a hydrolysate-based or a CD medium, used in the production of recombinantly-expressed proteins with sufficient galactose and/or galactose-containing supplement to achieve at least about the following galactose concentrations in the production media: at least about 1, at least about 4, at least about 5, at least about 10, at least about 15, at least about 20, at least about 30, at least about 40, at least about 60, or at least about 100 mM, wherein that production media is used to dilute a supplement-free cell culture growth media containing no supplement by a ratio of about 1:4 or about 1:5 (supplement-free growth media: supplemented production media).
  • In certain embodiments, the present invention is directed to methods of controlling the galactosylation profile of recombinantly-expressed proteins by supplementing a production medium, e.g., a hydrolysate-based or a CD medium, used in the production of recombinantly-expressed proteins with sufficient manganese and/or a manganese-containing supplement and sufficient galactose and/or galactose-containing supplement to achieve at least about the following manganese (Mn) and galactose (Gal) concentrations in the production media presented as Mn (μM)/Gal (mM): 0/1, 0/4, 0/5, 0/10, 0/15, 0/20, 0/30, 0/40, 0/60, 0/100, 0.1/0, 0.2/0, 0.5/0, 1.0/0, 10/0, 20/0, 25/0, 40/0, 50/0, 75/0, 80/0, 100/0, 0.2/1, 0.2/4, 0.2/30, 0.5/1, 0.5/4, 0.5/30, 10/10, 10/20, 10/40, 20/10, 20/20, 20/40, 25/15, 40/10, 40/20, 40/40, 40/100, 50/30, 60/20, 60/40, 60/100, 80/20, 80/40, 80/100, 100/20, 100/40, 100/100, wherein that production media is used to dilute a supplement-free cell culture growth media containing no supplement by a ratio of about 1:4 or about 1:5 (supplement-free growth media: supplemented production media).
  • In certain embodiments, the present invention is directed to methods of controlling the galactosylation profile of recombinantly-expressed proteins by supplementing a production medium, e.g., a hydrolysate-based or a CD medium, used in the production of recombinantly-expressed proteins with sufficient manganese and/or a manganese-containing supplement and sufficient galactose and/or galactose-containing supplement to achieve at least about the following manganese (Mn) and galactose (Gal) concentrations in the production media presented as Mn (μM)/Gal (mM): 0.2/1, 0.2/4, 0.2/30, 0.5/1, 0.5/4, 0.5/30, 10/10, 10/20, 10/40, 20/10, 20/20, 20/40, 25/15, 40/10, 40/20, 40/40, 40/100, 50/30, 60/20, 60/40, 60/100, 80/20, 80/40, 80/100, 100/20, 100/40, 100/100, wherein that production media is used to dilute a supplement-free cell culture growth media containing no supplement by a ratio of about 1:4 or about 1:5 (supplement-free growth media: supplemented production media).
  • In certain embodiments, the production medium, e.g., a hydrolysate-based or a CD medium, used in the production of a recombinantly-expressed protein is supplemented with manganese and not galactose. For the shake flasks studies in Example 1, but not by way of limitation, addition of manganese and not galactose to production IVGN CDM GIA-1 lowered the NGA2F+NGA2F-GlcNac sum by 6% to 9% and increased the NA1F+NA2F sum by 8% to 9% (FIGS. 3, 4, and 5). No further increase in manganese concentration was explored in the experimental design due to the growth inhibition observed at about 100 μM.
  • In certain embodiments, the production medium, e.g., a hydrolysate-based or a CD medium, used in the production of a recombinantly-expressed protein is supplemented with galactose and not manganese. For the shake flasks studies in Example 1, but not by way of limitation, addition of galactose only to production IVGN CDM GIA-1 lowered the NGA2F+NGA2F-GlcNac sum by 3% to 7% and increased NA1F+NA2F by 3% to 7% (FIGS. 3, 4, and 5). These findings indicate that a manganese concentration of about 100 μM and a galactose concentration of about 100 mM represent the maximum range of interest for this Example 1.
  • In certain embodiments, the production medium, e.g., a hydrolysate-based media or a CD media, used in the production of a recombinantly-expressed protein is supplemented with both manganese and galactose. For example, but not by way of limitation, the studies outlined in Example 1 indicate that the addition of combinations of manganese and galactose to production IVGN CDM GIA-1 resulted in a significant decrease in the NGA2F+NGA2F-GlcNac sum of 11% to 26% and a corresponding significant increase in the NA1F+NA2F sum of 13% to 23% as compared to the control condition where no manganese or galactose were added to the production media (FIGS. 3, 4, and 5). The effect on modulation of galactosylation of adalimumab in production IVGN CDM GIA-1 with the combined addition of manganese chloride and galactose was synergistic. In particular, the combined addition of manganese chloride and galactose decreased the NGA2F+NGA2F-GlcNac sum and increased the NA1F+NA2F sum by a larger percentage than by adding manganese or galactose alone and summing up their individual effects. For example, but not by way of limitation, addition of 40 μM manganese chloride alone reduced the NGA2F sum by 6%, and addition of 40 mM galactose alone decreased the NGA2F+NGA2F-GlcNac sum by 6%. However, the combined addition of manganese chloride and galactose at these same concentrations (i.e. 40 μM manganese+40 mM galactose) led to an 18% reduction in the NGA2F+NGA2F-GlcNac sum, 6% higher than their combined individual contributions to the reduction of the NGA2F+NGA2F-GlcNac sum. We define this effect as being synergistic and maintain this definition throughout the invention. The largest percent decrease in the NGA2F+NGA2F-GlcNac sum of approximately 26% was observed with the combined addition of 100 μM manganese chloride and 100 mM galactose. The largest percent increase in the NA1F+NA2F sum of approximately 23% was recorded with the combined addition of 60 μM manganese chloride and 100 mM galactose.
  • For the fed-batch bioreactor study described in Example 1, two manganese chloride and galactose combinations were studied and the results indicate that the decrease in the NGA2F+NGA2F-GlcNac sum and the corresponding increase in the NA1F+NA2F sum was scale (1.5 L vs. 200 mL) and process independent (fed-batch in controlled bioreactor environment vs. batch in shake flasks). For example, but not by way of limitation, the combined addition of 40 μM manganese chloride and 20 mM galactose to both production basal CDM GIA-1 and feed CDM JCL-5 decreased the NGA2F+NGA2F-GlcNac sum by 26% and increased the NA2F+NA2F sum by 27% compared to the control cultures (FIG. 6). A further increase in the galactose concentration to 40 mM in addition to manganese supplementation at 40 μM concentration resulted in an additional 3% decrease in the NGA2F+NGA2F-GlcNac sum, and a corresponding 3% increase in the NA1F+NA2F sum
  • In certain embodiments, the present invention is directed to the supplementation of CD media used in the production of a recombinantly-expressed protein with galactose and/or manganese. That such supplementation is effective across distinct CD media is evidenced by the results outlined in Example 2. Specifically, Example 2 results indicate that the addition of manganese chloride alone within the range of 0 to 40 μM to production CDM HyClone CDM4CHO decreased the NGA2F+NGA2F-GlcNac sum by a maximum of 5% in a concentration dependent manner (FIG. 8). A comparable maximum increase of 4% in the NA1F+NA2F sum was also achieved. Addition of galactose alone up to a maximum concentration of 40 mM yielded a 6% maximum decrease in the NGA2F+NGA2F-GlcNac sum and a corresponding 6% increase in the NA1F+NA2F sum. Modulation of galactosylation was also observed in production CDM HyClone CDM4CHO cultures supplemented with both manganese chloride and galactose. An additive effect was observed in cultures supplemented with both manganese chloride and galactose. The combined addition of manganese chloride and galactose decreased the NGA2F+NGA2F-GlcNac sum and increased the NA1F+NA2F sum by a comparable percentage as when manganese or galactose were added alone and their individual effects were summed up (FIG. 9). For example, but not by way of limitation, addition of 40 μM manganese chloride alone reduced the NGA2F+NGA2F-GlcNac sum by 5%, and addition of 40 mM galactose alone decreased the NGA2F sum by 6%. The combined addition of manganese chloride and galactose at these same concentrations (i.e. 40 μM manganese+40 mM galactose) led to a 12% reduction in the NGA2F+NGA2F-GlcNac sum. We define this effect as being additive and maintain this definition throughout the invention. The highest percentage decrease in the NGA2F sum of 12% and the corresponding 11% increase in the NA1F+NA2F sum was observed for the culture supplemented with 40 μM manganese chloride and 40 mM galactose.
  • In certain embodiments, the present invention is directed to the supplementation of a hydrolysate-based media used in the production of a recombinantly-expressed protein with galactose and/or manganese. For example, as outlined in Example 3, the addition of manganese chloride alone within the range of 0 to 40 μM to hydrolysate-based production media decreased the NGA2F+NGA2F-GlcNac sum by approximately 1%, although that change is within the oligosaccharide assay variability (FIG. 11). The addition of galactose alone up to a maximum concentration of 40 mM yielded a maximum decrease of 4% in the NGA2F+NGA2F-GlcNac sum and a corresponding 4% maximum increase in the NA1F+NA2F sum. Such oligosaccharide profile changes achieved with the addition of galactose alone are comparable to the changes recorded when combinations of galactose and manganese chloride were added to the hydrolysate-based media. For example, the combined addition of manganese chloride ranging from 0 to 40 μM and galactose ranging from 0 to 40 mM to hydrolysate-based media led to an approximate 5% maximum decrease in the NGA2F+NGA2F-GlcNac sum and a corresponding 3% increase in the NA1F+NA2F sum (FIG. 12). The highest percentage decrease of 5% in the NGA2F+NGA2F-GlcNac sum and the corresponding 4% increase in the NA1F+NA2F sum was observed for the culture supplemented with 40 mM galactose and either 20 μM or 40 μM manganese chloride.
  • The compositions and methods of the present invention also find use across distinct cell lines. For example, but not by way of limitation, the study described in Example 4 illustrates that the supplementation of a CD media, GIA-1, with galactose and/or manganese is effective to modulate galactosylation of adalimumab produced using a CHO cell line distinct from that employed in Examples 1-3. For example, but not by way of limitation, when using this alternative cell line, the addition of manganese chloride alone within the range of 0 to 20 μM to production CDM GIA-1 decreased the NGA2F+NGA2F-GlcNac sum in a concentration dependent manner and increased the NA1F+NA2F sum by approximately the same percentage. A maximum decrease of 22% in the NGA2F+NGA2F-GlcNac sum and a maximum corresponding increase of 23% in the NA1F+NA2F sum was observed with the addition of 20 μM manganese chloride (FIG. 14). Similarly, a concentration dependent decrease in the NGA2F+NGA2F-GlcNac sum and a corresponding increase in the NA1F+NA2F sum was observed with the addition of galactose alone in the range of 0 to 20 mM. A maximum decrease of 9% in the NGA2F+NGA2F-GlcNac sum and a corresponding maximum increase of 10% in the NA1F+NA2F sum was observed with the addition of 20 mM galactose. Similarly, an additive effect was observed for the oligosaccharide profiles of adalimumab produced in cultures supplemented with the combined addition of manganese chloride and galactose to GIA-1 media (FIG. 15). For example, but not by way of limitation, addition of 10 μM manganese chloride alone reduced the NGA2F+NGA2F-GlcNac sum by 18%, and addition of 10 mM galactose alone decreased the NGA2F+NGA2F-GlcNac sum by 6%. The combined addition of manganese chloride and galactose at these same concentrations led to a 24% reduction in the NGA2F+NGA2F-GlcNac sum. The highest percentage decrease of 35% in the NGA2F+NGA2F-GlcNac sum and the corresponding increase of 37% in the NA1F+NA2F sum were observed for the culture supplemented with 40 μM manganese chloride and 20 mM galactose.
  • That the compositions and methods of the present invention also find use across distinct cell lines is further reinforced by the results of Example 5, which employs a third adalimumab-producing cell line that is distinct from either of the adalimumab-producing cell lines of Examples 1-4. For example, but not by way of limitation, when using this third cell line, the addition of manganese chloride alone within the range of 0 to 1 μM to production CDM GIA-1 decreased the NGA2F+NGA2F-GlcNac sum in a concentration dependent manner and increased the NA1F+NA2F sum by approximately the same percentage. A maximum decrease of 26% in the NGA2F+NGA2F-GlcNac and a corresponding increase of 28% in the NA1F+NA2F oligosaccharides were observed with the addition of 1 μM manganese chloride (FIG. 17). The addition of galactose alone at 30 mM concentration to production CDM GIA-1 decreased the NGA2F+NGA2F-GlcNac sum by 4% and increased the NA1F+NA2F sum by 3%. Furthermore, when manganese chloride and galactose were supplemented together into the production basal and feed media, the results demonstrated a synergistic benefit towards the decrease in the NGA2F+NGA2F-GlcNAc and the increase in the NA1F+NA2F oligosaccharides which is consistent with the results demonstrated in Example 1 (FIG. 18). For example, but not by way of limitation, at 0.2 μM manganese chloride plus 30 mM galactose the observed 25% decrease in the NGA2F+NGA2F-GlcNAc sum was 6% more than the sum of the decrease observed with the addition of 0.2 μM manganese chloride alone (15%) and that of 30 mM galactose alone (4%). Similarly, the resulting 24% increase in the NA1F+NA2F sum was more than the sum of the increase observed with the addition of 0.2 μM manganese chloride alone (16%) and that of 30 mM galactose alone (3%). The combined supplementation of 0.5 μM manganese chloride+30 mM galactose also demonstrated a synergistic effect on the galactosylation profile of adalimumab produced in this third cell line. A maximum decrease compared to the control condition of 34% in the NGA2F+NGA2F-GlcNac and a corresponding 34% maximum increase in the NA1F+NA2F oligosaccharides was observed with the combined addition of 0.5 μM manganese chloride and 30 mM galactose to chemically defined GIA-1 media.
  • That the compositions and methods of the present invention also find use across distinct types of cell lines is further reinforced by the results of Example 6, which employs a fourth adalimumab-producing cell line that is distinct from the adalimumab-producing cell lines of Examples 1-5, in that it is an NSO cell line. For example, but not by way of limitation, when using this NSO cell line, the addition of manganese chloride alone within the range of 0 to 0.5 μM to production CDM PFBM-3/PFFM-4 decreased the NGA2F+NGA2F-GlcNac sum in a concentration dependent manner and increased the NA1F+NA2F sum by approximately the same percentage. A maximum decrease of 18% in the NGA2F+NGA2F-GlcNac sum and a corresponding increase of 20% in the NA1F+NA2F sum were observed with the addition of 0.5 μM manganese chloride (FIG. 20). However, manganese doses greater than 0.5 μM were not explored further due to cytotoxicity effects. Similarly, a concentration dependent decrease in the NGA2F+NGA2F-GlcNac sum and a corresponding increase in the NA1F+NA2F sum were observed with the addition of galactose alone in the range of 0 to 10 mM to production CDM PFBM-3/PFFM-4. A maximum decrease of 14% in the NGA2F+NGA2F-GlcNac sum and a corresponding increase of 15% in the NA1F+NA2F sum was observed with the addition of 10 mM galactose. In addition, the effect on modulation of galactosylation of adalimumab produced in a NSO cell line in production CDM PFBM-3/PFFM-4 supplemented with manganese chloride and galactose was synergistic (FIG. 21). For example, but not by way of limitation, addition of 0.2 μM manganese chloride alone reduced the NGA2F+NGA2F-GlcNac sum by 12%, and addition of 4 mM galactose alone decreased the NGA2F+NGA2F-GlcNac sum by 2%. However, the combined addition of manganese chloride and galactose at these same concentrations (i.e. 0.2 μM manganese+4 mM galactose) led to a 19% reduction in the NGA2F+NGA2F-GlcNac sum, 5% higher than their combined individual contributions. A maximum decrease of −26% in the NGA2F+NGA2F-GlcNac sum and a corresponding 28% increase in the NA1F+NA2F sum were observed with the combined addition of 0.5 μM manganese chloride and 4 mM galactose.
  • The compositions and methods of the present invention also find use in the production of diverse antibodies, as evidenced by the results of Example 7, which employs a CHO cell line that produced an antibody distinct from adalimumab. For example, but not by way of limitation, when producing this antibody distinct from adalimumab, the addition of manganese chloride alone within the range of 0 to 40 μM to production CDM GIA-1 decreased the NGA2F+NGA2F-GlcNac sum by a maximum of 26% (FIG. 23). A comparable maximum increase of 27% in the NA1F+NA2F sum was also achieved. Addition of galactose alone up to a maximum concentration of 40 mM yielded a maximum decrease of 12% in the NGA2F+NGA2F-GlcNac sum and a corresponding 13% maximum increase in the NA1F+NA2F sum in a concentration dependent manner. In addition, the combined addition of galactose and manganese chloride to production CDM GIA-1 resulted in a greater percent reduction in the NGA2F+NGA2F-GlcNac sum and, correspondingly, a greater percent increase in the NA1F+NA2F sum as compared to the addition of either component alone (FIG. 24). For example, but not by way of limitation, the addition of 40 μM manganese chloride alone reduced the NGA2F+NGA2F-GlcNac sum by 20%, and the addition of 40 mM galactose alone decreased the NGA2F+NGA2F-GlcNac sum by 12%. However, the combined addition of manganese chloride and galactose at these same concentrations (i.e. 40 μM manganese+40 mM galactose) led to a 27% decrease in the NGA2F+NGA2F-GlcNac sum. The highest percentage decrease of 32% in the NGA2F+NGA2F-GlcNac sum and the corresponding increase of 30% in the NA1F+NA2F sum were observed for the culture supplemented with 20 μM manganese chloride and 20 mM galactose.
  • That the compositions and methods of the present invention also find use when producing diverse antibodies is further reinforced by the results of Example 8, which employs a CHO cell line producing an antibody distinct from both adalimumab and the antibody of Example 7. For example, but not by way of limitation, when producing this third antibody, the addition of manganese chloride alone in the range of 0 to 75 μM to production CDM GIA-1 decreased the NGA2F+NGA2F-GlcNac sum by a maximum of 18% (FIG. 26). A comparable maximum increase of 16% in the NA1F+NA2F sum was also achieved. Addition of galactose alone up to a maximum concentration of 60 mM yielded a maximum decrease of 12% in the NGA2F+NGA2F-GlcNac sum and a corresponding 11% maximum increase in the NA1F+NA2F sum. In addition, when manganese chloride and galactose were supplemented together into the basal and feed media, the results demonstrated at least an additive effect and sometimes a synergistic effect towards the decrease in the NGA2F+NGA2F-GlcNAc sum and the increase in the NA1F+NA2F sum (FIG. 27). The synergistic effect was observed for the condition supplemented with 25 μM manganese chloride and 15 mM galactose. The observed 22% decrease in the NGA2F+NGA2F-GlcNAc sum was 5% more than the sum of the decrease observed with the addition of 25 μM manganese chloride alone (10%) and 15 mM galactose alone (7%). The additive effect was observed for the condition supplemented with 50 μM manganese chloride and 30 mM galactose. The observed 28% decrease in the NGA2F+NGA2F-GlcNAc sum was comparable to the sum of the decrease observed with the addition of 50 μM manganese chloride alone (18%) and 30 mM galactose alone (12%). A maximum decrease of 28% in the NGA2F+NGA2F-GlcNac and a corresponding 25% maximum increase in the NA1F+NA2F sum compared to the control condition was observed with the combined addition of 50 μM manganese chloride and 30 mM galactose to chemically defined GIA-1 media.
  • Although specifically directed to the production of antibodies, the following description outlines general techniques that can be adapted for the production of other recombinantly-expressed proteins. For example, to express a recombinant antibody, nucleic acids encoding partial or full-length light and heavy chains are inserted into one or more expression vector such that the genes are operatively linked to transcriptional and translational control sequences. (See, e.g., U.S. Pat. No. 6,914,128, the entire teaching of which is incorporated herein by reference.) In this context, the term “operatively linked” is intended to mean that an antibody gene is ligated into a vector such that transcriptional and translational control sequences within the vector serve their intended function of regulating the transcription and translation of the antibody gene. The expression vector and expression control sequences are chosen to be compatible with the expression host cell used. The antibody light chain gene and the antibody heavy chain gene can be inserted into a separate vector or, more typically, both genes are inserted into the same expression vector. The antibody genes are inserted into an expression vector by standard methods (e.g., ligation of complementary restriction sites on the antibody gene fragment and vector, or blunt end ligation if no restriction sites are present). Prior to insertion of the antibody or antibody-related light or heavy chain sequences, the expression vector may already carry antibody constant region sequences. For example, one approach to converting particular VH and VL sequences to full-length antibody genes is to insert them into expression vectors already encoding heavy chain constant and light chain constant regions, respectively, such that the VH segment is operatively linked to the CH segment(s) within the vector and the VL segment is operatively linked to the CL segment within the vector. Additionally or alternatively, the recombinant expression vector can encode a signal peptide that facilitates secretion of the antibody chain from a host cell. The antibody chain gene can be cloned into the vector such that the signal peptide is linked in-frame to the amino terminus of the antibody chain gene. The signal peptide can be an immunoglobulin signal peptide or a heterologous signal peptide (i.e., a signal peptide from a non-immunoglobulin protein).
  • In addition to the antibody chain genes, a recombinant expression vector of the invention can carry one or more regulatory sequence that controls the expression of the antibody chain genes in a host cell. The term “regulatory sequence” is intended to include promoters, enhancers and other expression control elements (e.g., polyadenylation signals) that control the transcription or translation of the antibody chain genes. Such regulatory sequences are described, e.g., in Goeddel; Gene Expression Technology: Methods in Enzymology 185, Academic Press, San Diego, Calif. (1990), the entire teaching of which is incorporated herein by reference. It will be appreciated by those skilled in the art that the design of the expression vector, including the selection of regulatory sequences may depend on such factors as the choice of the host cell to be transformed, the level of expression of protein desired, etc. Suitable regulatory sequences for mammalian host cell expression include viral elements that direct high levels of protein expression in mammalian cells, such as promoters and/or enhancers derived from cytomegalovirus (CMV) (such as the CMV promoter/enhancer), Simian Virus 40 (SV40) (such as the SV40 promoter/enhancer), adenovirus, (e.g., the adenovirus major late promoter (AdMLP)) and polyoma. For further description of viral regulatory elements, and sequences thereof, see, e.g., U.S. Pat. No. 5,168,062 by Stinski, U.S. Pat. No. 4,510,245 by Bell et al. and U.S. Pat. No. 4,968,615 by Schaffner et al., the entire teachings of which are incorporated herein by reference.
  • In addition to the antibody chain genes and regulatory sequences, a recombinant expression vector of the invention may carry one or more additional sequences, such as a sequence that regulates replication of the vector in host cells (e.g., origins of replication) and/or a selectable marker gene. The selectable marker gene facilitates selection of host cells into which the vector has been introduced (see e.g., U.S. Pat. Nos. 4,399,216, 4,634,665 and 5,179,017, all by Axel et al., the entire teachings of which are incorporated herein by reference). For example, typically the selectable marker gene confers resistance to drugs, such as G418, hygromycin or methotrexate, on a host cell into which the vector has been introduced. Suitable selectable marker genes include the dihydrofolate reductase (DHFR) gene (for use in dhfr-host cells with methotrexate selection/amplification) and the neo gene (for G418 selection).
  • An antibody, or antibody portion, of the invention can be prepared by recombinant expression of immunoglobulin light and heavy chain genes in a host cell. To express an antibody recombinantly, a host cell is transfected with one or more recombinant expression vectors carrying DNA fragments encoding the immunoglobulin light and heavy chains of the antibody such that the light and heavy chains are expressed in the host cell and secreted into the medium in which the host cells are cultured, from which medium the antibodies can be recovered. Standard recombinant DNA methodologies are used to obtain antibody heavy and light chain genes, incorporate these genes into recombinant expression vectors and introduce the vectors into host cells, such as those described in Sambrook, Fritsch and Maniatis (eds), Molecular Cloning; A Laboratory Manual, Second Edition, Cold Spring Harbor, N.Y., (1989), Ausubel et al. (eds.) Current Protocols in Molecular Biology, Greene Publishing Associates, (1989) and in U.S. Pat. Nos. 4,816,397 & 6,914,128, the entire teachings of which are incorporated herein.
  • For expression of the light and heavy chains, the expression vector(s) encoding the heavy and light chains is (are) transfected into a host cell by standard techniques. The various forms of the term “transfection” are intended to encompass a wide variety of techniques commonly used for the introduction of exogenous DNA into a prokaryotic or eukaryotic host cell, e.g., electroporation, calcium-phosphate precipitation, DEAE-dextran transfection and the like. Although it is theoretically possible to express the antibodies of the invention in either prokaryotic or eukaryotic host cells, expression of antibodies in eukaryotic cells, such as mammalian host cells, is suitable because such eukaryotic cells, and in particular mammalian cells, are more likely than prokaryotic cells to assemble and secrete a properly folded and immunologically active antibody. Prokaryotic expression of antibody genes has been reported to be ineffective for production of high yields of active antibody (Boss and Wood (1985) Immunology Today 6:12-13, the entire teaching of which is incorporated herein by reference).
  • Suitable host cells for cloning or expressing the DNA in the vectors herein are the prokaryote, yeast, or higher eukaryote cells described above. Suitable prokaryotes for this purpose include eubacteria, such as Gram-negative or Gram-positive organisms, e.g., Enterobacteriaceae such as Escherichia, e.g., E. coli, Enterobacter, Erwinia, Klebsiella, Proteus, Salmonella, e.g., Salmonella typhimurium, Serratia, e.g., Serratia marcescans, and Shigella, as well as Bacilli such as B. subtilis and B. licheniformis (e.g., B. licheniformis 41P disclosed in DD 266,710 published Apr. 12, 1989), Pseudomonas such as P. aeruginosa, and Streptomyces. One suitable E. coli cloning host is E. coli 294 (ATCC 31,446), although other strains such as E. coli B, E. coli X1776 (ATCC 31,537), and E. coli W3110 (ATCC 27,325) are suitable. These examples are illustrative rather than limiting.
  • In addition to prokaryotes, eukaryotic microbes such as filamentous fungi or yeast are suitable cloning or expression hosts for polypeptide encoding vectors. Saccharomyces cerevisiae, or common baker's yeast, is the most commonly used among lower eukaryotic host microorganisms. However, a number of other genera, species, and strains are commonly available and useful herein, such as Schizosaccharomyces pombe; Kluyveromyces hosts such as, e.g., K. lactis, K. fragilis (ATCC 12,424), K. bulgaricus (ATCC 16,045), K. wickeramii (ATCC 24,178), K. waltii (ATCC 56,500), K. drosophilarum (ATCC 36,906), K. thermotolerans, and K. marxianus; yarrowia (EP 402,226); Pichia pastoris (EP 183,070); Candida; Trichoderma reesia (EP 244,234); Neurospora crassa; Schwanniomyces such as Schwanniomyces occidentalis; and filamentous fungi such as, e.g., Neurospora, Penicillium, Tolypocladium, and Aspergillus hosts such as A. nidulans and A. niger.
  • Suitable host cells for the expression of glycosylated antibodies are derived from multicellular organisms. Examples of invertebrate cells include plant and insect cells. Numerous baculoviral strains and variants and corresponding permissive insect host cells from hosts such as Spodoptera frugiperda (caterpillar), Aedes aegypti (mosquito), Aedes albopictus (mosquito), Drosophila melanogaster (fruitfly), and Bombyx mori have been identified. A variety of viral strains for transfection are publicly available, e.g., the L-1 variant of Autographa californica NPV and the Bm-5 strain of Bombyx mori NPV, and such viruses may be used as the virus herein according to the present invention, particularly for transfection of Spodoptera frugiperda cells. Plant cell cultures of cotton, corn, potato, soybean, petunia, tomato, and tobacco can also be utilized as hosts.
  • Suitable mammalian host cells for expressing the recombinant antibodies of the invention include Chinese Hamster Ovary (CHO cells) (including dhfr-CHO cells, described in Urlaub and Chasin, (1980) PNAS USA 77:4216-4220, used with a DHFR selectable marker, e.g., as described in Kaufman and Sharp (1982) Mol. Biol. 159:601-621, the entire teachings of which are incorporated herein by reference), NS0 myeloma cells, COS cells and SP2 cells. When recombinant expression vectors encoding antibody genes are introduced into mammalian host cells, the antibodies are produced by culturing the host cells for a period of time sufficient to allow for expression of the antibody in the host cells or secretion of the antibody into the culture medium in which the host cells are grown. Other examples of useful mammalian host cell lines are monkey kidney CV1 line transformed by SV40 (COS-7, ATCC CRL 1651); human embryonic kidney line (293 or 293 cells subcloned for growth in suspension culture, Graham et al., J. Gen Virol. 36:59 (1977)); baby hamster kidney cells (BHK, ATCC CCL 10); Chinese hamster ovary cells/-DHFR (CHO, Urlaub et al., Proc. Natl. Acad. Sci. USA 77:4216 (1980)); mouse sertoli cells (TM4, Mather, Biol. Reprod. 23:243-251 (1980)); monkey kidney cells (CV1 ATCC CCL 70); African green monkey kidney cells (VERO-76, ATCC CRL-1587); human cervical carcinoma cells (HELA, ATCC CCL 2); canine kidney cells (MDCK, ATCC CCL 34); buffalo rat liver cells (BRL 3A, ATCC CRL 1442); human lung cells (W138, ATCC CCL 75); human liver cells (Hep G2, HB 8065); mouse mammary tumor (MMT 060562, ATCC CCL51); TRI cells (Mather et al., Annals N.Y. Acad. Sci. 383:44-68 (1982)); MRC 5 cells; FS4 cells; and a human hepatoma line (Hep G2), the entire teachings of which are incorporated herein by reference.
  • Host cells are transformed with the above-described expression or cloning vectors for antibody production and cultured in conventional nutrient media modified as appropriate for inducing promoters, selecting transformants, or amplifying the genes encoding the desired sequences.
  • The host cells used to produce an antibody may be cultured in a variety of media. Commercially available media such as Ham's F10™ (Sigma), Minimal Essential Medium™ ((MEM), (Sigma), RPMI-1640 (Sigma), and Dulbecco's Modified Eagle's Medium™ ((DMEM), Sigma) are suitable for culturing the host cells. In addition, any of the media described in Ham et al., Meth. Enz. 58:44 (1979), Barnes et al., Anal. Biochem. 102:255 (1980), U.S. Pat. Nos. 4,767,704; 4,657,866; 4,927,762; 4,560,655; or 5,122,469; WO 90/03430; WO 87/00195; or U.S. Pat. No. Re. 30,985 may be used as culture media for the host cells, the entire teachings of which are incorporated herein by reference. The culture conditions, such as temperature, pH, and the like, are those previously used with the host cell selected for expression, and will be apparent to the ordinarily skilled artisan.
  • Host cells can also be used to produce portions of intact antibodies, such as Fab fragments or scFv molecules. It is understood that variations on the above procedure are within the scope of the present invention. For example, in certain embodiments it may be desirable to transfect a host cell with DNA encoding either the light chain or the heavy chain (but not both) of an antibody of this invention. Recombinant DNA technology may also be used to remove some or all of the DNA encoding either or both of the light and heavy chains that is not necessary for antigen binding. The molecules expressed from such truncated DNA molecules are also encompassed by the antibodies of the invention. In addition, bifunctional antibodies may be produced in which one heavy and one light chain are an antibody of the invention and the other heavy and light chain are specific for an antigen other than the original antigen by crosslinking an antibody of the invention to a second antibody by standard chemical crosslinking methods.
  • In a suitable system for recombinant expression of an antibody, or antigen-binding portion thereof, of the invention, a recombinant expression vector encoding both the antibody heavy chain and the antibody light chain is introduced into dhfr-CHO cells by calcium phosphate-mediated transfection. Within the recombinant expression vector, the antibody heavy and light chain genes are each operatively linked to CMV enhancer/AdMLP promoter regulatory elements to drive high levels of transcription of the genes. The recombinant expression vector also carries a DHFR gene, which allows for selection of CHO cells that have been transfected with the vector using methotrexate selection/amplification. The selected transformant host cells are cultured to allow for expression of the antibody heavy and light chains and intact antibody is recovered from the culture medium. Standard molecular biology techniques are used to prepare the recombinant expression vector, transfect the host cells, select for transformants, culture the host cells and recover the antibody from the culture medium.
  • When using recombinant techniques, the antibody can be produced intracellularly, in the periplasmic space, or directly secreted into the medium. In one aspect, if the antibody is produced intracellularly, as a first step, the particulate debris, either host cells or lysed cells (e.g., resulting from homogenization), can be removed, e.g., by centrifugation or ultrafiltration. Where the antibody is secreted into the medium, supernatants from such expression systems can be first concentrated using a commercially available protein concentration filter, e.g., an Amicon™ or Millipore Pellicon™ ultrafiltration unit.
  • Prior to the process of the invention, procedures for purification of antibodies from cell debris initially depend on the site of expression of the antibody. Some antibodies can be secreted directly from the cell into the surrounding growth media; others are made intracellularly. For the latter antibodies, the first step of a purification process typically involves: lysis of the cell, which can be done by a variety of methods, including mechanical shear, osmotic shock, or enzymatic treatments. Such disruption releases the entire contents of the cell into the homogenate, and in addition produces subcellular fragments that are difficult to remove due to their small size. These are generally removed by differential centrifugation or by filtration. Where the antibody is secreted, supernatants from such expression systems are generally first concentrated using a commercially available protein concentration filter, e.g., an Amicon™ or Millipore Pellicon™ ultrafiltration unit. Where the antibody is secreted into the medium, the recombinant host cells can also be separated from the cell culture medium, e.g., by tangential flow filtration. Antibodies can be further recovered from the culture medium using the antibody purification methods of the invention.
  • 6. EXAMPLES 6.1. Example 1 6.1.1. Materials & Methods
  • In the studies summarized in this example, we investigated the effects on product quality attributes resulting from the addition of manganese chloride and/or galactose to chemically defined Life Technologies Gibco, GIA-1, media (proprietary formulation) in the adalimumab-producing CHO cell line utilized in Example 3, but adapted to GIA-1 media. The studies were performed in either a batch process in shake flasks or a fed-batch process in 3 L bioreactors.
  • Growth and production media for the adalimumab-producing CHO cell line were prepared using a proprietary Life Technologies Gibco chemically defined media, GIA-1. Basal production and feed media were supplemented with Manganese (II) Chloride (Sigma M1787-100 mL; 1.0 M±0.1 M) and D(+)Galactose (Sigma G5388-1 kg) according to the experimental design described in Table 1. All media were filtered through Corning 0.5 L or 1 L filter systems 0.22 μm Poly(Ether Sulfone) (PES) and stored at 4° C. until use.
  • The cell line utilized for both studies was generated from the adalimumab-producing CHO cell utilized in Example 3 by adapting it to chemically defined GIA-1 media for 7 (2 to 3 day each) passages in a combination of 250 mL and 500 mL Corning vented non-baffled shake flasks before freezing.
  • Upon thaw, for the batch shake flask study, cells were expanded for 3 to 5 (2 to 3 day each) passages in a combination of 250 mL and 500 mL Corning vented non-baffled shake flasks. Production cultures were initiated in duplicate 500 mL Corning vented non-baffled shake flasks (200 mL working volume) at an initial viable cell density (VCD) of approximately 0.5×106 cells/mL. Cultures were maintained on orbital shakers at 110 revolutions per minute (RPM) in a dry incubator at 35° C. and 5% CO2. The shake flask study was run in an extended batch mode by feeding a glucose solution (1.25% (v/v) of 40% solution) when the media glucose concentration fell below 3 g/L.
  • For the fed-batch bioreactor study, cells were expanded for 8 (2 to 3 day each) passages in Corning vented non-baffled shake flasks maintained on orbital shakers at 110 RPM and in 20 L cell bags (3 L to 10 L working volume) maintained at 20-25 RPM, 7.5° angle, and 0.25 SLPM airflow in a dry incubator at 35° C. and 5% CO2. Production cultures were initiated in duplicate 3 L bioreactors (1.5 L working volume) at 35° C., 30% dissolved oxygen, 200 RPM, pH ramp from 7.1 to 6.9 over 3 days, and pH setpoint of 6.9 thereafter. A fixed split ratio of cells to media of 1:5 was utilized to initiate the production stage cultures. In the fed-batch mode, a chemically-defined feed from Life Technologies Gibco, JCL-5 (proprietary formulation), was added as follows: 3% (v/v)— day 3, 5%— day 4, 7%— day 5, 10%— day 6, and 10%—day 7. Additional glucose (1.25% (v/v) of 40% solution) was fed when the media glucose concentration fell below 3 g/L.
  • For all studies with CHO cell lines described throughout this invention, samples were collected daily and measured for cell density and viability using a Cedex cell counter. Retention samples for titer analysis via Poros A method were collected by centrifugation at 12,000 RPM for 5 min when the culture viability began declining. The cultures were harvested by collecting 125 mL aliquots and centrifuging at 3,000 RPM for 30 min when culture viability was near or below 50%. All supernatants were stored at −80° C. until analysis.
  • For all studies, the harvest samples were Protein A purified and prepared for the oligosaccharide assay using the following procedures. As a first step in the process of establishing the identity and quantifying the oligosaccharides, they are released from the protein by enzymatic digestion with N-glycanase. Once the glycans are released, the free reducing end of each glycan is labeled by reductive amination with a fluorescent tag, 2-aminobenzamide (2-AB). The resulting labeled glycans are separated by normal-phase HPLC (NP-HPLC) in acetonitrile: 50 mM ammonium formate, pH 4.4, and detected by a fluorescence detector. Quantitation is based on the relative area percent of detected sugars. Throughout this invention, the relative area percentages of the agalactosyl fucosylated biantennary oligosaccharides, denoted as NGA2F+NGA2F-GlcNAc sum, and the galactose-containing fucosylated biantennary oligosaccharides NA1F+NA2F sum are reported and discussed.
  • 6.1.2. Experimental Design
  • As detailed in Table 1, for the batch shake flask study, manganese chloride was supplemented at the following concentrations in production media: 0, 40, 60, 80, and 100 μM. Galactose was supplemented at the following levels in production media: 0, 10, 20, 40, and 100 mM. Individual and combined additions of manganese chloride and galactose were studied using a comprehensive design divided into 3 sets of experiments. Each experiment had a control culture for direct comparison of culture growth, productivity, and product quality. Production media used for the control cultures was not supplemented with manganese chloride or galactose. Culture growth, productivity, and product quality data for control cultures is the average of the 3 experiments.
  • For the fed-batch bioreactor study, manganese chloride and galactose were supplemented to both production and feed media in the following combinations: 40 μM manganese chloride and 20 mM galactose; 40 μM manganese chloride and 40 mM galactose (Table 2). Basal and feed media for the control cultures were not supplemented with manganese chloride or galactose.
  • TABLE 1
    Experimental design for the
    batch shake flasks study (Example 1)
    Manganese Galactose
    (μM) (mM) ID
    0 0 Mn(0) Gal(0)
    0 10 Mn(0) Gal(10)
    0 20 Mn(0) Gal(20)
    0 40 Mn(0) Gal(40)
    0 100 Mn(0) Gal(100)
    40 0 Mn(40) Gal (0)
    80 0 Mn(80) Gal(0)
    100 0 Mn(100) Gal(0)
    40 10 Mn(40) Gal(10)
    40 20 Mn(40) Gal(20)
    40 40 Mn(40) Gal(40)
    40 100 Mn(40) Gal(100)
    60 20 Mn(60) Gal(20)
    60 40 Mn(60) Gal(40)
    60 100 Mn(60) Gal(100)
    80 20 Mn(80) Gal(20)
    80 40 Mn(80) Gal(40)
    80 100 Mn(80) Gal(100)
    100 20 Mn(100) Gal(20)
    100 40 Mn(100) Gal(40)
    100 100 Mn(100) Gal(100)
  • TABLE 1
    Experimental design for the fed-batch
    3L bioreactors study (Example 1)
    Manganese Galactose
    (μM) (mM) ID
    0 0 Mn(0) Gal(0)
    40 20 Mn(40) Gal(20)
    40 40 Mn(40) Gal(40)
  • 6.1.3. Culture Growth & Productivity
  • For the shake flasks experiments, cell growth and viability profiles of cultures in production media supplemented with galactose in the 0 to 100 mM concentration range and/or manganese chloride up to 80 μM concentration were comparable to control cultures without manganese and/or galactose added (FIGS. 1A, 1B). Cultures grown in media supplemented with manganese chloride at 100 μM concentration and galactose concentrations in the 0 to 100 mM range experienced growth lag and decreased viability for the first 4 production days, likely due to toxic effects of manganese at this concentration. However, toxicity effects were overcome after production day 4. Harvest titer for most experimental conditions was 3% to 14% higher than the average harvest titer for the control cultures (FIG. 1C). All three control cultures had comparable growth profiles and productivity. For the bioreactors fed-batch experiment, culture growth, viability profiles, and harvest titer were comparable for all conditions (FIGS. 2A, 2B, 2C).
  • 6.1.4. Oligosaccharide Analysis
  • In this example, the modulation of galactosylation with the addition of manganese chloride and/or galactose to chemically defined media GIA-1 was explored using the adalimumab-producing CHO cell line utilized in Example 3, but adapted to GIA-1 media.
  • 6.1.4.1. Batch Shake Flask Study
  • For the shake flask study, the addition of manganese chloride alone within the range of 0 to 100 μM to production CDM GIA-1 decreased the NGA2F+NGA2F-GlcNac sum in a concentration dependent manner and increased the NA1F+NA2F sum by approximately the same percentage. A maximum change of 9% in the NGA2F+NGA2F-GlcNac and 8% in the NA1F+NA2F oligosaccharides was observed with the addition of 100 μM manganese chloride (FIG. 3). Manganese doses greater than 100 μM were not explored further due to cytotoxicity effects. Similarly, a concentration dependent decrease in the NGA2F+NGA2F-GlcNac sum and a corresponding increase in the NA1F+NA2F sum were observed with the addition of galactose alone in the range of 0 to 100 mM to production CDM GIA-1. A maximum change of 7% in the NGA2F+NGA2F-GlcNac and the NA1F+NA2F oligosaccharides was observed with the addition of 100 mM galactose.
  • The effect on modulation of galactosylation of adalimumab in CDM GIA-1 with the combined addition of manganese chloride and galactose was synergistic. Combined addition of manganese chloride and galactose decreased the NGA2F+NGA2F-GlcNac sum and increased the NA1F+NA2F sum by a larger percentage than by adding manganese or galactose alone and summing up their individual effects (FIGS. 4 and 5). For example, addition of 40 μM manganese chloride alone reduced the NGA2F+NGA2F-GlcNac sum by 6%, and addition of 40 mM galactose alone decreased the NGA2F+NGA2F-GlcNac sum by 6%. However, the combined addition of manganese chloride and galactose at these same concentrations (i.e. 40 μM manganese+40 mM galactose) led to an 18% reduction in the NGA2F+NGA2F-GlcNac sum, 6% higher than their combined individual contributions to the reduction of the NGA2F+NGA2F-GlcNac sum. We define this effect as being synergistic and maintain this definition throughout the invention. A maximum decrease compared to the control condition of 26% in the NGA2F+NGA2F-GlcNac sum was observed with the combined addition of 100 μM manganese chloride and 100 mM galactose.
  • 6.1.4.2. Fed-Batch Bioreactor Study
  • For the fed-batch 3 L bioreactors study, two manganese chloride and galactose combinations were studied and we show that the decrease in the NGA2F+NGA2F-GlcNac and the corresponding increase in the NA1F+NA2F oligosaccharides was scale (1.5 L vs. 200 mL) and process independent (fed-batch in controlled bioreactor environment vs. batch in shake flasks). Combined addition of 40 μM manganese chloride and 20 mM galactose to both production basal CDM GIA-1 and feed CDM JCL-5 decreased the NGA2F+NGA2F-GlcNac sum by 26% and increased the NA2F+NA2F sum by 27% compared to the control cultures (FIG. 6). A further increase in the galactose concentration to 40 mM in addition to manganese supplementation at 40 μM concentration resulted in an additional 3% decrease in the NGA2F+NGA2F-GlcNac sum, and a corresponding 3% increase in the NA1F+NA2F sum.
  • 6.2. Example 2 6.2.1. Materials & Methods
  • In the study summarized in this example, we investigated the effects on product quality attributes resulting from the addition of manganese chloride and/or galactose to the chemically defined media Thermo Scientific HyClone CDM4CHO using the adalimumab-producing CHO cell line used in Example 1 further adapted to HyClone media.
  • Growth and production media for the adalimumab-producing CHO cell line were prepared using Thermo Scientific HyClone chemically defined media CDM4CHO without L-glutamine (Catalogue # SH30558.02). Production media was supplemented with Manganese (II) Chloride (Sigma M1787-100 mL; 1.0 M±0.1 M) and D(+)Galactose (Sigma G5388-1 kg) according to the experimental design described in Table 3. All media were filtered through Corning 0.5 L or 1 L (0.22 μm PES) filter systems and stored at 4° C. until use.
  • Upon thaw, cells were adapted to and expanded in HyClone CDM4CHO media for 5 (2 to 3 day each) passages in a combination of 250 mL, 500 mL, and 1000 mL Corning vented non-baffled shake flasks. Production cultures were initiated in duplicate 500 mL Corning vented non-baffled shake flasks (200 mL working volume) at an initial VCD of approximately 0.5×106 cells/mL. Cultures were maintained on orbital shakers at 110 RPM in a dry incubator at 35° C. and 5% CO2. A glucose solution (1.25% (v/v) of 40% solution) was fed when the media glucose concentration fell below 3 g/L.
  • 6.2.2. Experimental Design
  • As detailed in Table 3, manganese chloride was supplemented at the following concentrations in production media: 0, 10, 20, and 40 μM. Galactose was supplemented at the following levels in production media: 0, 10, 20, and 100 mM. Production media for the control cultures was not supplemented with manganese chloride or galactose.
  • TABLE 3
    Experimental design for Example 2
    Manganese Galactose
    (μM) (mM) ID
    0 0 Mn(0) Gal(0)
    0 10 Mn(0) Gal(10)
    0 20 Mn(0) Gal(20)
    0 40 Mn(0) Gal(40)
    10 0 Mn(10) Gal (0)
    20 0 Mn(20) Gal(0)
    40 0 Mn(40) Gal(0)
    10 10 Mn(10) Gal(10)
    20 20 Mn(20) Gal(20)
    40 40 Mn(40) Gal(40)
  • 6.2.3. Culture Growth & Productivity
  • Cell growth and viability profiles of cultures in production HyClone CDM4CHO supplemented with galactose in the 0 to 40 mM concentration range and/or manganese chloride up to 10 μM concentration were comparable to the control cultures without manganese and/or galactose added (FIGS. 7A, 7B). Increasing the concentration of manganese chloride in HyClone CDM4CHO production media to 20 μM or 40 μM slowed down culture growth. Manganese doses greater than 40 μM were not explored further due to the observed growth inhibition effects. Harvest titer for all conditions was comparable to the control (data not shown).
  • 6.2.4. Oligosaccharide Analysis
  • In this example, the modulation of galactosylation with the addition of manganese chloride and/or galactose to the commercially available HyClone CDM4CHO was explored using the adalimumab-producing CHO cell line used Example 1 further adapted to HyClone media.
  • The addition of manganese chloride alone within the range of 0 to 40 μM to production CDM HyClone CDM4CHO decreased the NGA2F+NGA2F-GlcNac sum by a maximum of 5% in a concentration dependent manner (FIG. 8). A comparable maximum increase of 4% in the NA1F+NA2F sum was achieved. Addition of galactose alone up to a maximum concentration of 40 mM yielded a 6% maximum decrease in the NGA2F+NGA2F-GlcNac sum and a corresponding 6% increase in the NA1F+NA2F sum.
  • An additive effect was observed in cultures supplemented with both manganese chloride and galactose. The combined addition of manganese chloride and galactose decreased the NGA2F+NGA2F-GlcNac sum and increased the NA1F+NA2F sum by a comparable percentage as when manganese or galactose were added alone and their individual effects were summed up (FIG. 9). For example, addition of 40 μM manganese chloride alone reduced the NGA2F+NGA2F-GlcNac sum by 5%, and addition of 40 mM galactose alone decreased the NGA2F+NGA2F-GlcNac sum by 6%. The combined addition of manganese chloride and galactose at these same concentrations (i.e. 40 μM manganese+40 mM galactose) led to a 12% reduction in the NGA2F+NGA2F-GlcNac sum. We define this effect as being additive and maintain this definition throughout the invention.
  • The highest percentage decrease in the NGA2F+NGA2F-GlcNac sum of 12% and the corresponding 11% increase in the NA1F+NA2F sum was observed for the culture supplemented with 40 μM manganese chloride and 40 mM galactose.
  • 6.3. Example 3 6.3.1. Materials & Methods
  • In the study summarized in this example, we investigated the effects on product quality attributes resulting from the addition of manganese chloride and/or galactose to a hydrolysate-based media (proprietary formulation) in an adalimumab-producing CHO cell line.
  • Growth and production media for the adalimumab-producing CHO cell line were prepared using yeast and soy hydrolysates according to a proprietary formulation. Production media was supplemented with Manganese (II) Chloride (Sigma M1787-100 mL; 1.0 M±0.1 M) and D(+)Galactose (Sigma G5388-1 kg) according to the experimental design described in Table 4. All media were filtered through Corning 0.5 L or 1 L filter systems (0.22 μm PES) and stored at 4° C. until use.
  • Upon thaw, cells were expanded for 9 (2 to 3 day each) passages in a combination of 250 mL, 500 mL, and 1000 mL Corning vented non-baffled shake flasks. Production cultures were initiated in duplicate 500 mL Corning vented non-baffled shake flasks (200 mL working volume) at an initial VCD of approximately 0.5×106 cells/mL. Cultures were maintained on orbital shakers at 110 RPM in a dry incubator at 35° C. and 5% CO2. A glucose solution (1.25% (v/v) of 40% solution) was fed when the media glucose concentration fell below 3 g/L.
  • 6.3.2. Experimental Design
  • As detailed in Table 4, manganese chloride was supplemented at the following concentrations in production media: 0, 10, 20, and 40 μM. Galactose was supplemented at the following levels in production media: 0, 10, 20, and 40 mM. Production media for the control cultures was not supplemented with manganese chloride or galactose.
  • TABLE 4
    Experimental design for Example 3
    Manganese Galactose
    (μM) (mM) ID
    0 0 Mn(0) Gal(0)
    0 10 Mn(0) Gal(10)
    0 20 Mn(0) Gal(20)
    0 40 Mn(0) Gal(40)
    10 0 Mn(10) Gal (0)
    20 0 Mn(20) Gal(0)
    40 0 Mn(40) Gal(0)
    10 10 Mn(10) Gal(10)
    20 20 Mn(20) Gal(20)
    40 40 Mn(40) Gal(40)
  • 6.3.3. Culture Growth & Productivity
  • Cell growth of most cultures in the hydrolysate-based media supplemented with galactose in the 0 to 40 mM concentration range and/or manganese chloride in the 0 to 40 μM concentration was slower compared to the control cultures without manganese or galactose added (FIG. 10A). However, all cultures reached a comparable peak viable cell density. Some cultures supplemented with 20 μM or 40 μM manganese chloride showed decreased viability by day 3 of production culture, but recovered as the cultures progressed (FIG. 10B). The culture supplemented with 10 μM manganese chloride was studied with a second control condition (B) in a separate experiment. Both these cultures grew to slightly higher maximum VCD compared to all other cultures, however results were within historical variation. Harvest titer for all conditions was comparable to the control (data not shown).
  • 6.3.4. Oligosaccharide Analysis
  • In this example, the modulation of galactosylation by the addition of manganese chloride and/or galactose to a hydrolysate-based media was explored using an adalimumab-producing CHO cell line in shake flasks.
  • The addition of manganese chloride alone within the range of 0 to 40 μM to hydrolysate-based production media decreased the NGA2F+NGA2F-GlcNac sum by approximately 1%, a change that is within the oligosaccharide assay variability (FIG. 11). The addition of galactose alone up to a maximum concentration of 40 mM yielded a maximum decrease of 4% in the NGA2F+NGA2F-GlcNac sum and a corresponding 4% maximum increase in the NA1F+NA2F sum.
  • The oligosaccharide profile changes achieved with the addition of galactose alone are comparable to the changes recorded when combinations of galactose and manganese chloride were added to the hydrolysate-based media. The combined addition of manganese chloride ranging from 0 to 40 μM and galactose ranging from 0 to 40 mM to hydrolysate-based media led to an approximate 5% maximum decrease in the NGA2F+NGA2F-GlcNac sum and a corresponding 3% increase in the NA1F+NA2F sum (FIG. 12). The highest percentage decrease of 5% in the NGA2F+NGA2F-GlcNac sum and the corresponding 4% increase in the NA1F+NA2F sum was observed for the culture supplemented with 40 mM galactose and either 20 μM or 40 μM manganese chloride.
  • 6.4. Example 4 6.4.1. Materials & Methods
  • In the study summarized in this example, we investigated the effects on product quality attributes resulting from the addition of manganese chloride and/or galactose to chemically defined Life Technologies Gibco GIA-1 media using a different adalimumab producing CHO cell line than in Examples 1, 2, and 3, named CHO cell line #2.
  • Growth and production media for the adalimumab-producing CHO cell line #2 were prepared using a proprietary Life Technologies Gibco chemically defined media, GIA-1. Production media only was supplemented with Manganese (II) Chloride (Sigma M1787-100 mL; 1.0 M±0.1 M) and D(+)Galactose (Sigma G5388-1 kg) according to the experimental design described in Table 5. All media were filtered through Corning 0.5 L or 1 L filter systems (0.22 μm PES) and stored at 4° C. until use.
  • Upon thaw, cells were expanded for 5 to 8 (2 to 3 day each) passages in a combination of 250 mL, 500 mL, and 1000 mL Corning vented non-baffled shake flasks. Production cultures were initiated in duplicate 500 mL Corning vented non-baffled shake flasks (200 mL working volume) at an initial VCD of approximately 0.5×106 cells/mL. Cultures were maintained on orbital shakers at 180 RPM in a dry incubator at 35° C. and 5% CO2. A glucose solution (1.25% (v/v) of 40% solution) was fed when the media glucose concentration fell below 3 g/L.
  • 6.4.2. Experimental Design
  • As detailed in Table 5, manganese chloride was supplemented at the following concentrations in production media: 0, 10, 20, and 40 μM. Galactose was supplemented at the following levels in production media: 0, 10, and 20 mM. Production media for the control cultures was not supplemented with manganese chloride or galactose. This study was run in 2 blocks.
  • TABLE 5
    Experimental design for Example 4
    Manganese Galactose
    (μM) (mM) ID
    Block I 0 0 Mn(0) Gal(0)
    20 0 Mn(20) Gal(0)
    10 10 Mn(10) Gal(10)
    20 20 Mn(20) Gal(20)
    40 20 Mn(40) Gal(20)
    Block II 0 0 Mn(0) Gal(0)
    0 10 Mn(0) Gal(10)
    0 20 Mn(0) Gal (20)
    10 0 Mn(10) Gal(0)
  • 6.4.3. Culture Growth & Productivity
  • Culture growth, viability profiles, and harvest titer of cultures in production CDM GIA-1 supplemented with galactose in the 0 to 20 mM concentration range and/or manganese chloride in the 0 to 40 μM concentration range were comparable to the control cultures without manganese or galactose added (FIGS. 13A, 13B; harvest titer data not shown).
  • 6.4.4. Oligosaccharide Analysis
  • In this example, the modulation of galactosylation by the addition of manganese chloride and/or galactose to chemically defined GIA-1 media was explored using the adalimumab-producing CHO cell line #2.
  • The addition of manganese chloride alone within the range of 0 to 20 μM to production CDM GIA-1 decreased the NGA2F+NGA2F-GlcNac sum in a concentration dependent manner and increased the NA1F+NA2F sum by approximately the same percentage. A maximum decrease of 22% in the NGA2F+NGA2F-GlcNac sum and a maximum corresponding increase of 23% in the NA1F+NA2F sum was observed with the addition of 20 μM manganese chloride (FIG. 14). Similarly, a concentration dependent decrease in the NGA2F+NGA2F-GlcNac sum and a corresponding increase in the NA1F+NA2F sum was observed with the addition of galactose alone in the range of 0 to 20 mM. A maximum decrease of 9% in the NGA2F+NGA2F-GlcNac sum and a corresponding maximum increase of 10% in the NA1F+NA2F sum was observed with the addition of 20 mM galactose.
  • An additive effect was observed for the oligosaccharide profiles of adalimumab produced in cultures supplemented with the combined addition of manganese chloride and galactose to GIA-1 media (FIG. 15). For example, addition of 10 μM manganese chloride alone reduced the NGA2F+NGA2F-GlcNac sum by 18%, and addition of 10 mM galactose alone decreased the NGA2F+NGA2F-GlcNac sum by 6%. The combined addition of manganese chloride and galactose at these same concentrations led to a 24% reduction in the NGA2F+NGA2F-GlcNac sum. The highest percentage decrease of 35% in the NGA2F+NGA2F-GlcNac sum and the corresponding increase of 37% in the NA1F+NA2F sum were observed for the culture supplemented with 40 μM manganese chloride and 20 mM galactose.
  • 6.5. Example 5 6.5.1. Materials & Methods
  • In the study summarized in this example, we investigated the effects on product quality attributes resulting from the addition of manganese chloride and/or galactose to chemically defined Life Technologies Gibco GIA-1 media using a different adalimumab-producing CHO cell line than in Examples 1, 2, 3, and 4, named CHO cell line #3.
  • Growth and production media for the adalimumab-producing CHO cell line #3 were prepared using the proprietary Life Technologies Gibco chemically defined media, GIA-1. Basal production and feed media were supplemented with Manganese (II) Chloride (Sigma M3634-100 g) and D(+)Galactose (Sigma G5388-1 kg) according to the experimental design described in Table 6.
  • Upon thaw, cells were expanded in Corning vented non-baffled shake flasks maintained on orbital shakers at 140 RPM, and in 10 L cell bags (2 L working volume) maintained at 25 RPM, 7° angle, and 0.25 SLPM airflow in a dry incubator at 36° C. and 5% CO2. Production cultures were initiated in 3 L bioreactors (1.4 L initial working volume) at 36° C., 30% dissolved oxygen, 200 RPM, and pH 6.9±0.2. A fixed split ratio of cells to media of 1:6.7 was utilized to initiate the production stage cultures. A temperature shift was performed when the culture VCD reached a value higher than 5×106 cells/mL. The chemically-defined feed from Life Technologies Gibco JCL-5 was added as follows: 4% (v/v)— day 2, 6%— day 4, 8%— day 6, 10%— day 8, and 10%—day 10. Additional 400 g/L glucose was added to the reactor cultures as needed to ensure glucose levels did not deplete. Bioreactors were harvested at a viability of approximately 50% or on production day 17, whichever condition occurred first.
  • 6.5.2. Experimental Design
  • As detailed in Table 6, manganese chloride was supplemented at the following concentrations in both production and feed media: 0, 0.1, 0.2, 0.5, and 1.0 μM. Galactose was supplemented at 0 and 30 mM concentrations in both production and feed media. In addition, a combined manganese chloride and galactose supplementation strategy was utilized for the production basal and feed media at either 0.2 or 0.5 μM manganese chloride plus 30 mM galactose. Basal and feed media for the control cultures were not supplemented with manganese chloride or galactose.
  • TABLE 6
    Experimental design for Example 5
    Manganese Galactose
    (μM) (mM) ID
    0 0 Mn(0) Gal(0)
    0.1 0 Mn(0.1) Gal(0)
    0.2 0 Mn(0.2) Gal(0)
    0.5 0 Mn(0.5) Gal(0)
    1.0 0 Mn(1.0) Gal(0)
    0 30 Mn(0) Gal(30)
    0.2 30 Mn(0.2) Gal(30)
    0.5 30 Mn(0.5) Gal(30)
  • 6.5.3. Culture Growth & Productivity
  • Growth profiles of most cultures supplemented with manganese chloride and/or galactose were comparable to the control culture except for the cultures supplemented with 30 mM galactose alone or in combination with 0.2 μM manganese chloride which grew slower and reached a lower peak VCD (FIG. 16A). However, the culture supplemented with 0.5 μM manganese chloride and 30 mM galactose had a growth profile comparable to the control culture indicating that neither manganese chloride nor galactose at the concentrations studied are detrimental to culture growth. Viability profiles and harvest titer were comparable to the control condition (FIGS. 16B, 16C).
  • 6.5.4. Oligosaccharide Analysis
  • In this example, the modulation of galactosylation with the addition of manganese chloride and/or galactose to chemically defined media GIA-1 was explored using the adalimumab-producing CHO cell line #3.
  • The addition of manganese chloride alone within the range of 0 to 1 μM to production CDM GIA-1 decreased the NGA2F+NGA2F-GlcNac sum in a concentration dependent manner and increased the NA1F+NA2F sum by approximately the same percentage. A maximum decrease of 26% in the NGA2F+NGA2F-GlcNac and a corresponding increase of 28% in the NA1F+NA2F oligosaccharides were observed with the addition of 1 μM manganese chloride (FIG. 17). The addition of galactose alone at 30 mM concentration to production CDM GIA-1 decreased the NGA2F+NGA2F-GlcNac sum by 4% and increased the NA1F+NA2F sum by 3%.
  • When manganese chloride and galactose were supplemented together into the production basal and feed media, the results demonstrated a synergistic benefit towards the decrease in the NGA2F+NGA2F-GlcNAc and the increase in the NA1F+NA2F oligosaccharides which is consistent with the results demonstrated in Example 1 (FIG. 18). For example, at 0.2 μM manganese chloride plus 30 mM galactose the observed 25% decrease in the NGA2F+NGA2F-GlcNAc sum was 6% more than the sum of the decrease observed with the addition of 0.2 μM manganese chloride alone (15%) and that of 30 mM galactose alone (4%). Similarly the resulting 24% increase in the NA1F+NA2F sum was more than the sum of the increase observed with the addition of 0.2 μM manganese chloride alone (16%) and that of 30 mM galactose alone (3%). The combined supplementation of 0.5 μM manganese chloride+30 mM galactose also demonstrated a synergistic effect on the galactosylation profile of adalimumab produced in the CHO cell line #3. A maximum decrease compared to the control condition of 34% in the NGA2F+NGA2F-GlcNac and a corresponding 34% maximum increase in the NA1F+NA2F oligosaccharides was observed with the combined addition of 0.5 μM manganese chloride and 30 mM galactose to chemically defined GIA-1 media.
  • 6.6. Example 6 6.6.1. Materials & Methods
  • In the study summarized in this example, we investigated the effects on product quality attributes resulting from the addition of manganese chloride and/or galactose to chemically defined Life Technologies Gibco PFBM-3 basal medium and PFFM-4 feed medium (proprietary formulation) using an adalimumab-producing NSO cell line in a fed-batch process in shake flasks.
  • Growth and production media for the adalimumab-producing NSO cell line were prepared using a proprietary Life Technologies Gibco chemically defined media, PFBM-3 basal medium plus PFFM-4 feed medium. Production and feed media were supplemented with Manganese (II) Chloride (Sigma M1787-100 mL; 1.0 M±0.1 M) and D(+)Galactose (Sigma G5388-1 kg) according to the experimental design described in Table 7.
  • Upon thaw, cells were expanded for 3 to 5 (2 days each) passages in a combination of 250 mL, 500 mL, 1 L, 2 L and 3 L Corning vented non-baffled shake flasks. Production cultures were initiated in single 1 L Corning vented non-baffled shake flasks (240 mL initial working volume) at an initial VCD of approximately 2.5×105 cells/mL. Cultures were maintained on orbital shakers at 100 RPM in a dry incubator at 37° C. and 5% CO2. The shake flask study was run in a fed-batch mode and the culture was fed PFFM-4 as follows: 24 mL—day 2, 28.8 mL—day 4, 28.8 mL—day 6, and 28.8 mL—day 8.
  • Samples were collected every 2 days and measured for cell density and viability using a Cedex cell counter. Retention samples for titer analysis via Poros A method were collected daily beginning on Day 8 by centrifugation at 2,000 g for 10 min and then filtered through 0.2 um PVDF syringe filter. The cultures were harvested on production day 10. The entire culture was collected, chilled on ice to approximately 0° C. for 1.5 hours, the cells and debris flocculated at pH 5.0 by the addition of 1M citric acid and held for 15 minutes, and centrifuged at 4000×g for 15 min at 5° C. The supernatant was passed through 0.20 um Millipore Stericup PES filters, and, immediately post filtration, the acidified clarified cell-free harvest was neutralized with 2M Tris to pH 7.1±0.2. The cell free harvest was transferred to PETG bottles and stored at −80° C. until analysis.
  • 6.6.2. Experimental Design
  • As detailed in Table 7, manganese chloride was supplemented at the following concentrations in both production and feed media: 0, 0.2, and 0.5 μM. Galactose was supplemented at the following levels in both production and feed media: 0, 1, 4, 5, and 10 mM. Manganese chloride and galactose were added in a full factorial, two level DOE design for the 0, 1, and 4 mM galactose conditions and all concentrations of manganese chloride. Individual and combined additions of manganese chloride and galactose were studied using a comprehensive design divided into 2 sets of experiments. Each experiment had a control culture for direct comparison of culture growth, productivity, and product quality. Production medium for control cultures was not supplemented with manganese chloride or galactose.
  • TABLE 7
    Experimental design for Example 6
    Manganese Galactose
    (μM) (mM) ID
    Block I 0 0 Mn(0) Gal(0)
    0 5 Mn(0) Gal(5)
    0 10 Mn(0) Gal(10)
    Block II 0 0 Mn(0) Gal(0)
    0.2 0 Mn(0.2) Gal(0)
    0.5 0 Mn(0.5) Gal(0)
    0 1 Mn(0) Gal(1)
    0.2 1 Mn(0.2) Gal(1)
    0.5 1 Mn(0.5) Gal(1)
    0 4 Mn(0) Gal(4)
    0.2 4 Mn(0.2) Gal(4)
    0.5 4 Mn(0.5) Gal(4)
  • 6.6.3. Culture Growth & Productivity
  • Culture growth and viability profiles in production media supplemented with galactose in the 0 to 5 mM concentration range and/or manganese chloride up to 0.5 μM concentration were comparable to the control condition without manganese or galactose added (FIGS. 19A, 19B). The addition of galactose at 10 mM concentration had a detrimental effect on culture growth and productivity. The cultures in the Block I experiment had a lower maximum VCD and overall lower viability than the cultures in the Block II experiment. All cultures in the Block II experiment showed similar VCD and viability profiles. Harvest titer for most experimental conditions was comparable to the harvest titer for the control condition except for the titer of the culture supplemented with 10 mM galactose, which was 60% lower (FIG. 19C).
  • 6.6.4. Oligosaccharide Analysis
  • In this example, the modulation of galactosylation by the addition of manganese chloride and/or galactose to chemically defined PFBM-3/PFFM-4 media was explored using an adalimumab-producing NSO cell line in a fed-batch process in shake flasks.
  • The addition of manganese chloride alone within the range of 0 to 0.5 μM to CDM PFBM-3/PFFM-4 decreased the NGA2F+NGA2F-GlcNac sum in a concentration dependent manner and increased the NA1F+NA2F sum by approximately the same percentage. A maximum decrease of 18% in the NGA2F+NGA2F-GlcNac sum and a corresponding increase of 20% in the NA1F+NA2F sum were observed with the addition of 0.5 μM manganese chloride (FIG. 20). Manganese doses greater than 0.5 μM were not explored further due to cytotoxicity effects. Similarly, a concentration dependent decrease in the NGA2F+NGA2F-GlcNac sum and a corresponding increase in the NA1F+NA2F sum were observed with the addition of galactose alone in the range of 0 to 10 mM to CDM PFBM-3/PFFM-4. A maximum decrease of 14% in the NGA2F+NGA2F-GlcNac sum and a corresponding increase of 15% in the NA1F+NA2F sum was observed with the addition of 10 mM galactose.
  • The effect on modulation of galactosylation of adalimumab produced in a NSO cell line in CDM PFBM-3/PFFM-4 supplemented with manganese chloride and galactose was synergistic (FIG. 21). For example, addition of 0.2 μM manganese chloride alone reduced the NGA2F+NGA2F-GlcNac sum by 12%, and addition of 4 mM galactose alone decreased the NGA2F+NGA2F-GlcNac sum by 2%. However, the combined addition of manganese chloride and galactose at these same concentrations (i.e. 0.2 μM manganese+4 mM galactose) led to a 19% reduction in the NGA2F+NGA2F-GlcNac sum, 5% higher than their combined individual contributions. A maximum decrease of −26% in the NGA2F+NGA2F-GlcNac sum and a corresponding 28% increase in the NA1F+NA2F sum were observed with the combined addition of 0.5 μM manganese chloride and 4 mM galactose.
  • 6.7. Example 7 6.7.1. Materials & Methods
  • In the study summarized in this example, we investigated the effects on product quality attributes resulting from the addition of manganese chloride and/or galactose to chemically defined Life Technologies Gibco GIA-1 media in a CHO cell line producing a monoclonal antibody generically named mAb #1.
  • Growth and production media for the CHO cell line producing mAb #1 were prepared using a proprietary Life Technologies Gibco chemically defined media, GIA-1. Production media only was supplemented with Manganese (II) Chloride (Sigma M1787-100 mL; 1.0 M±0.1 M) and D(+)Galactose (Sigma G5388-1 kg) according to the experimental design described in Table 8. All media were filtered through Corning 0.5 L or 1 L filter systems (0.22 μm PES) and stored at 4° C. until use.
  • Upon thaw, cells were expanded for 6 (3 day each) passages in a combination of 250 mL, 500 mL, and 1000 mL Corning vented non-baffled shake flasks. Production cultures were initiated in duplicate 500 mL Corning vented non-baffled shake flasks (200 mL working volume) at an initial VCD of approximately 0.5×106 cells/mL. Cultures were maintained on orbital shakers at 125 RPM in a dry incubator at 35° C. and 5% CO2. A glucose solution (1.25% (v/v) of 40% solution) was fed when the media glucose concentration fell below 3 g/L.
  • 6.7.2. Experimental Design
  • As detailed in Table 8, manganese chloride was supplemented at the following levels in production media: 0, 10, 20, and 40 μM. Galactose was supplemented at the following levels in production media: 0, 10, 20, and 100 mM. Production media for the control cultures was not supplemented with manganese chloride or galactose.
  • TABLE 8
    Experimental design for Example 7
    Manganese Galactose
    (μM) (mM) ID
    0 0 Mn(0) Gal(0)
    0 10 Mn(0) Gal(10)
    0 20 Mn(0) Gal(20)
    0 40 Mn(0) Gal(40)
    10 0 Mn(10) Gal (0)
    20 0 Mn(20) Gal(0)
    40 0 Mn(40) Gal(0)
    10 10 Mn(10) Gal(10)
    20 20 Mn(20) Gal(20)
    40 40 Mn(40) Gal(40)
  • 6.7.3. Culture Growth & Productivity
  • Cultures supplemented with manganese chloride alone in the concentration range of 0 to 20 μM grew comparable to the control cultures (FIG. 22A). Cultures supplemented with galactose alone or with the combination of 20 μM manganese and 20 mM galactose grew to a lower maximum VCD compared to the control, but had the same growth rate until the peak VCD was achieved on production day 6. These cultures ended a day earlier, on production day 9 (FIG. 22B). Cultures supplemented with 40 μM manganese chloride and galactose at all levels studied along with the culture supplemented with 10 μM manganese chloride and 10 mM galactose experienced slower growth and decreased peak VCD compared to the control. Harvest titer was 3-24% lower than the control condition (data not shown).
  • 6.7.4. Oligosaccharide Analysis
  • In this example, the modulation of galactosylation by the addition of manganese chloride and/or galactose to chemically defined Life Technologies Gibco GIA-1 media was explored using a CHO cell line producing the monoclonal antibody mAb #1.
  • The addition of manganese chloride alone within the range of 0 to 40 μM to production CDM GIA-1 decreased the NGA2F+NGA2F-GlcNac sum by a maximum of 26% (FIG. 23). A comparable maximum increase of 27% in the NA1F+NA2F sum was achieved. Addition of galactose alone up to a maximum concentration of 40 mM yielded a maximum decrease of 12% in the NGA2F+NGA2F-GlcNac sum and a corresponding 13% maximum increase in the NA1F+NA2F sum in a concentration dependent manner.
  • The combined addition of galactose and manganese chloride to production CDM GIA-1 resulted in a greater percent reduction in the NGA2F+NGA2F-GlcNac sum and, correspondingly, a greater percent increase in the NA1F+NA2F sum as compared to the addition of either component alone (FIG. 24). For example, the addition of 40 μM manganese chloride alone reduced the NGA2F+NGA2F-GlcNac sum by 20%, and the addition of 40 mM galactose alone decreased the NGA2F+NGA2F-GlcNac sum by 12%. However, the combined addition of manganese chloride and galactose at these same concentrations (i.e. 40 μM manganese+40 mM galactose) led to a 27% decrease in the NGA2F+NGA2F-GlcNac sum. The highest percentage decrease of 32% in the NGA2F+NGA2F-GlcNac sum and the corresponding increase of 30% in the NA1F+NA2F sum were observed for the culture supplemented with 20 μM manganese chloride and 20 mM galactose.
  • 6.8. Example 8 6.8.1. Materials & Methods
  • In the study summarized in this example, we investigated the effects on product quality attributes resulting from the addition of manganese chloride and/or galactose to chemically defined Life Technologies Gibco GIA-1 media in a CHO cell line producing the monoclonal antibody generically named mAb #2 in a fed-batch process in 3 L bioreactors.
  • Growth and production media for the mAb #2 producing CHO cell line were prepared using the proprietary Life Technologies Gibco chemically defined media, GIA-1. Basal production and feed media were supplemented with Manganese (II) Chloride (Sigma M3634) and D(+)Galactose (Sigma G5388-1 kg) according to the experimental design described in Table 9.
  • Upon thaw, cells were expanded in Corning vented non-baffled shake flasks maintained on orbital shakers at 140 RPM, and in 10 L cell bags (2 L working volume) maintained at 25 RPM, 7° angle, 0.25 SLPM airflow in a dry incubator at 36° C. and 5% CO2. Production cultures were initiated in 3 L bioreactors (1.5 L initial working volume) at 36° C., 25% dissolved oxygen, 200 RPM, and pH 7.0. The chemically-defined feed from Life Technologies Gibco JCL-5 was added as follows: 3% (v/v)— day 3, 5%— day 5, 7%— day 7, 10%— day 9, and 10%—day 11. Additional 400 g/L glucose was added to the bioreactor cultures as needed to ensure the glucose levels did not deplete. Bioreactors were harvested at viability of approximately 70% or below or on production day 15, whichever condition occurred first.
  • 6.8.2. Experimental Design
  • As detailed in Table 9, manganese chloride was supplemented at the following concentrations in both production and feed media: 0, 25, 50, and 75 μM. Galactose was supplemented at 0, 15, 30, and 60 mM concentrations in both production and feed media. In addition, a combined manganese chloride and galactose supplementation strategy was utilized for the production basal and feed media at 25 μM manganese chloride+15 mM galactose, and 50 μM manganese chloride+30 mM galactose. Basal and feed media for the control cultures were not supplemented with manganese chloride or galactose.
  • TABLE 9
    Experimental design for Example 8
    Manganese Galactose
    (μM) (mM) ID
    0 0 Mn(0) Gal(0)
    25 0 Mn(25) Gal(0)
    50 0 Mn(50) Gal(0)
    75 0 Mn(75) Gal(0)
    0 15 Mn(0) Gal(15)
    0 30 Mn(0) Gal(30)
    0 60 Mn(0) Gal(60)
    25 15 Mn(25) Gal(15)
    50 30 Mn(50) Gal(30)
  • 6.8.3. Culture Growth & Productivity
  • Growth profiles of most cultures supplemented with galactose in the 0 to 60 mM concentration range and/or manganese chloride in the 0 to 75 μM range were comparable to the control culture except for the culture supplemented with 25 μM manganese chloride alone which grew slower after production day 7 (FIG. 25A). However, increasing the amount of manganese chloride supplemented to production CDM GIA-1 to 50 μM or 75 μM resulted in cultures with growth profiles comparable to the control culture. Viability profiles and harvest titer were comparable to the control condition (FIGS. 25B, 25C).
  • 6.8.4. Oligosaccharide Analysis
  • In this example, the modulation of galactosylation with the addition of manganese chloride and/or galactose to chemically defined media GIA-1 was explored using a CHO cell line producing the monoclonal antibody mAb #2.
  • The addition of manganese chloride alone in the range of 0 to 75 μM to production CDM GIA-1 decreased the NGA2F+NGA2F-GlcNac sum by a maximum of 18% (FIG. 26). A comparable maximum increase of 16% in the NA1F+NA2F sum was achieved. Addition of galactose alone up to a maximum concentration of 60 mM yielded a maximum decrease of 12% in the NGA2F+NGA2F-GlcNac sum and a corresponding 11% maximum increase in the NA1F+NA2F sum.
  • When manganese chloride and galactose were supplemented together into the basal and feed media, the results demonstrated at least an additive effect and sometimes a synergistic effect towards the decrease in the NGA2F+NGA2F-GlcNAc and the increase in the NA1F+NA2F oligosaccharides (FIG. 27). The synergistic effect was observed for the condition supplemented with 25 μM manganese chloride and 15 mM galactose. The observed 22% decrease in the NGA2F+NGA2F-GlcNAc sum was 5% more than the sum of the decrease observed with the addition of 25 μM manganese chloride alone (10%) and 15 mM galactose alone (7%). The additive effect was observed for the condition supplemented with 50 μM manganese chloride and 30 mM galactose. The observed 28% decrease in the NGA2F+NGA2F-GlcNAc sum was comparable to the sum of the decrease observed with the addition of 50 μM manganese chloride alone (18%) and 30 mM galactose alone (12%). A maximum decrease of 28% in the NGA2F+NGA2F-GlcNac and a corresponding 25% maximum increase in the NA1F+NA2F sum compared to the control condition was observed with the combined addition of 50 μM manganese chloride and 30 mM galactose to chemically defined GIA-1 media.
  • All patents, patent applications, publications, product descriptions and protocols, cited in this specification are hereby incorporated by reference in their entirety. In case of a conflict in terminology, the present disclosure controls.
  • While it will be apparent that the invention herein described is well calculated to achieve the benefits and advantages set forth above, the present invention is not to be limited in scope by the specific embodiments described herein. It will be appreciated that the invention is susceptible to modification, variation and change without departing from the spirit thereof.

Claims (28)

What is claimed is:
1. A composition comprising an antibody comprising the heavy and light chain variable domains of adalimumab, wherein more than 25% of the total N-linked oligosaccharides present on said antibody are of a galactose-containing fucosylated biantennary oligosaccharide form (sum of NA1F+NA2F).
2. The composition of claim 1, wherein the antibody is adalimumab.
3. The composition of claim 1, wherein 26-40% of the total N-linked oligosaccharides present on said antibody are of a galactose-containing fucosylated biantennary oligosaccharide form (sum of NA1F+NA2F).
4. The composition of claim 1, wherein 35-44% of the total N-linked oligosaccharides present on said antibody are of a galactose-containing fucosylated biantennary oligosaccharide form (sum of NA1F+NA2F).
5. The composition of claim 1, wherein 30-40% of the total N-linked oligosaccharides present on said antibody are of a galactose-containing fucosylated biantennary oligosaccharide form (sum of NA1F+NA2F).
6. The composition of claim 1, wherein less than 70% of the total N-linked oligosaccharides present on said antibody are of an agalactosyl fucosylated biantennary oligosaccharide form (sum NGA2F+NGA2F-GlcNAc).
7. The composition of claim 6, wherein 49-69% of the total N-linked oligosaccharides present on said antibody are of an agalactosyl fucosylated biantennary oligosaccharide form (sum NGA2F+NGA2F-GlcNAc).
8. The composition of claim 6, wherein 55-65% of the total N-linked oligosaccharides present on said antibody are of an agalactosyl fucosylated biantennary oligosaccharide form (sum NGA2F+NGA2F-GlcNAc).
9. The composition of claim 1, wherein said antibody has been produced by culturing a mammalian cell in a cell culture media which has been supplemented with a manganese supplement and a galactose supplement.
10. The composition of claim 1, wherein said antibody has been produced by culturing a mammalian cell in a cell culture media comprising 0.2-100 μM of a manganese supplement and 1-100 mM of a galactose supplement.
11. The composition of claim 10, wherein the cell culture media comprises a hydrolysate based media or a chemically defined media.
12. The composition of claim 10, wherein the mammalian cell is a CHO cell or an NS0 cell.
13. A composition comprising an antibody comprising the heavy and light chain variable domains of adalimumab, wherein less than 70% of the total N-linked oligosaccharides present on said antibody are of a galactose-containing fucosylated biantennary oligosaccharide form (sum of NA1F+NA2F).
14. The composition of claim 13, wherein the antibody is adalimumab.
15. The composition of claim 13, wherein 49-69% of the total N-linked oligosaccharides present on said antibody are of an agalactosyl fucosylated biantennary oligosaccharide form (sum NGA2F+NGA2F-GlcNAc).
16. The composition of claim 13, wherein 55-65% of the total N-linked oligosaccharides present on said antibody are of an agalactosyl fucosylated biantennary oligosaccharide form (sum NGA2F+NGA2F-GlcNAc).
17. The composition of claim 13, wherein said antibody has been produced by culturing a mammalian cell in a cell culture media which has been supplemented with a manganese supplement and a galactose supplement.
18. The composition of claim 17, wherein the cell culture media has been supplemented with 0.2-100 μM of a manganese supplement and 1-100 mM of a galactose supplement.
19. The composition of claim 17, wherein the cell culture media comprises a hydrolysate based media or a chemically defined media.
20. The composition of claim 17, wherein the mammalian cell is a CHO cell or an NSO cell.
21. A composition comprising an antibody comprising the heavy and light chain variable domains of adalimumab, wherein less than 10% of the total N-linked oligosaccharides present on said antibody are of a galactose-containing fucosylated biantennary oligosaccharide form (sum of NA1F+NA2F)
22. The composition of claim 21, wherein the antibody is adalimumab.
23. The composition of claim 21, wherein 3-9.9% of the total N-linked oligosaccharides present on said antibody are of a galactose-containing fucosylated biantennary oligosaccharide form (sum of NA1F+NA2F).
24. The composition of claim 23, wherein more than 81% of the total N-linked oligosaccharides present on said antibody are of an agalactosyl fucosylated biantennary oligosaccharide form (sum NGA2F+NGA2F-GlcNAc).
25. The composition of claim 23, wherein said antibody has been produced by culturing a mammalian cell in a cell culture media which has been supplemented with a manganese supplement and a galactose supplement.
26. The composition of claim 25, wherein the cell culture media has been supplemented with 0.2-100 μM of a manganese supplement and 1-100 mM of a galactose supplement.
27. The composition of claim 25, wherein the cell culture media comprises a hydrolysate based media or a chemically defined media.
28. The composition of claim 25, wherein the mammalian cell is a CHO cell or an NSO cell.
US14/619,799 2011-04-27 2015-02-11 Methods for controlling the galactosylation profile of recombinantly-expressed proteins Active US9255143B2 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
US14/619,799 US9255143B2 (en) 2011-04-27 2015-02-11 Methods for controlling the galactosylation profile of recombinantly-expressed proteins
US15/014,694 US9365645B1 (en) 2011-04-27 2016-02-03 Methods for controlling the galactosylation profile of recombinantly-expressed proteins
US15/086,739 US9505834B2 (en) 2011-04-27 2016-03-31 Methods for controlling the galactosylation profile of recombinantly-expressed proteins
US15/345,953 US20170051052A1 (en) 2011-04-27 2016-11-08 Methods for controlling the galactosylation profile of recombinantly-expressed proteins

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201161479727P 2011-04-27 2011-04-27
US13/457,020 US9062106B2 (en) 2011-04-27 2012-04-26 Methods for controlling the galactosylation profile of recombinantly-expressed proteins
US14/493,068 US9090688B2 (en) 2011-04-27 2014-09-22 Methods for controlling the galactosylation profile of recombinantly-expressed proteins
US14/619,799 US9255143B2 (en) 2011-04-27 2015-02-11 Methods for controlling the galactosylation profile of recombinantly-expressed proteins

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US14/493,068 Continuation US9090688B2 (en) 2011-04-27 2014-09-22 Methods for controlling the galactosylation profile of recombinantly-expressed proteins

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US15/014,694 Division US9365645B1 (en) 2011-04-27 2016-02-03 Methods for controlling the galactosylation profile of recombinantly-expressed proteins

Publications (2)

Publication Number Publication Date
US20150158944A1 true US20150158944A1 (en) 2015-06-11
US9255143B2 US9255143B2 (en) 2016-02-09

Family

ID=46046346

Family Applications (6)

Application Number Title Priority Date Filing Date
US13/457,020 Active US9062106B2 (en) 2011-04-27 2012-04-26 Methods for controlling the galactosylation profile of recombinantly-expressed proteins
US14/493,068 Active US9090688B2 (en) 2011-04-27 2014-09-22 Methods for controlling the galactosylation profile of recombinantly-expressed proteins
US14/619,799 Active US9255143B2 (en) 2011-04-27 2015-02-11 Methods for controlling the galactosylation profile of recombinantly-expressed proteins
US15/014,694 Active US9365645B1 (en) 2011-04-27 2016-02-03 Methods for controlling the galactosylation profile of recombinantly-expressed proteins
US15/086,739 Active US9505834B2 (en) 2011-04-27 2016-03-31 Methods for controlling the galactosylation profile of recombinantly-expressed proteins
US15/345,953 Abandoned US20170051052A1 (en) 2011-04-27 2016-11-08 Methods for controlling the galactosylation profile of recombinantly-expressed proteins

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US13/457,020 Active US9062106B2 (en) 2011-04-27 2012-04-26 Methods for controlling the galactosylation profile of recombinantly-expressed proteins
US14/493,068 Active US9090688B2 (en) 2011-04-27 2014-09-22 Methods for controlling the galactosylation profile of recombinantly-expressed proteins

Family Applications After (3)

Application Number Title Priority Date Filing Date
US15/014,694 Active US9365645B1 (en) 2011-04-27 2016-02-03 Methods for controlling the galactosylation profile of recombinantly-expressed proteins
US15/086,739 Active US9505834B2 (en) 2011-04-27 2016-03-31 Methods for controlling the galactosylation profile of recombinantly-expressed proteins
US15/345,953 Abandoned US20170051052A1 (en) 2011-04-27 2016-11-08 Methods for controlling the galactosylation profile of recombinantly-expressed proteins

Country Status (4)

Country Link
US (6) US9062106B2 (en)
EP (1) EP2702077A2 (en)
TW (2) TW201708539A (en)
WO (1) WO2012149197A2 (en)

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9150645B2 (en) 2012-04-20 2015-10-06 Abbvie, Inc. Cell culture methods to reduce acidic species
US9193787B2 (en) 2012-04-20 2015-11-24 Abbvie Inc. Human antibodies that bind human TNF-alpha and methods of preparing the same
US9200070B2 (en) 2013-10-18 2015-12-01 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same
US9234033B2 (en) 2012-09-02 2016-01-12 Abbvie, Inc. Methods to control protein heterogeneity
US9249182B2 (en) 2012-05-24 2016-02-02 Abbvie, Inc. Purification of antibodies using hydrophobic interaction chromatography
US9255143B2 (en) 2011-04-27 2016-02-09 Abbvie Inc. Methods for controlling the galactosylation profile of recombinantly-expressed proteins
US9499614B2 (en) 2013-03-14 2016-11-22 Abbvie Inc. Methods for modulating protein glycosylation profiles of recombinant protein therapeutics using monosaccharides and oligosaccharides
US9499616B2 (en) 2013-10-18 2016-11-22 Abbvie Inc. Modulated lysine variant species compositions and methods for producing and using the same
US9550826B2 (en) 2013-11-15 2017-01-24 Abbvie Inc. Glycoengineered binding protein compositions
US9598667B2 (en) 2013-10-04 2017-03-21 Abbvie Inc. Use of metal ions for modulation of protein glycosylation profiles of recombinant proteins
US9688752B2 (en) 2013-10-18 2017-06-27 Abbvie Inc. Low acidic species compositions and methods for producing and using the same using displacement chromatography
US9708400B2 (en) 2012-04-20 2017-07-18 Abbvie, Inc. Methods to modulate lysine variant distribution
US9708399B2 (en) 2013-03-14 2017-07-18 Abbvie, Inc. Protein purification using displacement chromatography

Families Citing this family (37)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10059770B2 (en) 2012-01-30 2018-08-28 Dr. Reddy's Laboratories Limited Process of modulating man5 and/or afucosylation content of a glycoprotein composition
AU2013309506A1 (en) 2012-09-02 2015-03-12 Abbvie Inc. Methods to control protein heterogeneity
EP2931749B1 (en) * 2012-12-17 2019-04-24 Laboratoire Francais du Fractionnement et des Biotechnologies Societe Anonyme Use of monoclonal antibodies for the treatment of inflammation and bacterial infections
WO2014159579A1 (en) 2013-03-14 2014-10-02 Abbvie Inc. MUTATED ANTI-TNFα ANTIBODIES AND METHODS OF THEIR USE
TWI625390B (en) 2013-03-14 2018-06-01 安美基公司 Methods for increasing mannose content of recombinant proteins
JP6689189B2 (en) 2013-03-26 2020-04-28 コヒラス・バイオサイエンシズ・インコーポレイテッド Protein production method
US20160237399A1 (en) 2015-02-18 2016-08-18 Biogen Ma Inc. Control of Protein Glycosylation by Culture Medium Supplementation and Cell Culture Process Parameters
WO2015026846A1 (en) * 2013-08-19 2015-02-26 Biogen Idec Ma Inc. Control of protein glycosylation by culture medium supplementation and cell culture process parameters
CN106170298B (en) 2013-10-16 2024-01-09 前瞻疗法公司 Buffer formulations for improving antibody stability
US8946395B1 (en) 2013-10-18 2015-02-03 Abbvie Inc. Purification of proteins using hydrophobic interaction chromatography
AU2015211514B2 (en) 2014-01-29 2018-05-10 Lg Chem, Ltd. Method for modulating galactosylation of recombinant protein through optimization of culture medium
EP3110941A4 (en) 2014-02-25 2017-10-18 Dr. Reddy's Laboratories Ltd. Process for modifying galactosylation and g0f content of a glycoprotein composition by glutamine supplementation
KR102280638B1 (en) 2014-02-27 2021-07-22 에프. 호프만-라 로슈 아게 Modulation of cell growth and glycosylation in recombinant glycoprotein production
KR101660580B1 (en) * 2014-04-02 2016-09-28 프레스티지 바이오파마 피티이. 엘티디. A method for preparing an antibody by controlling a sugar content of the antibody
WO2016007764A1 (en) * 2014-07-09 2016-01-14 Abbvie Inc. Methods for modulating the glycosylation profile of recombinant proteins using non-commonly used sugars
CN108064266A (en) 2014-07-21 2018-05-22 格利科斯芬兰公司 The preparation of the glycoprotein with mammal sample N- glycan in filamentous fungi
MA41685A (en) * 2014-10-17 2017-08-22 Biogen Ma Inc COPPER SUPPLEMENT FOR THE REGULATION OF GLYCOSYLATION IN A MAMMAL CELL CULTURE PROCESS
US20160115225A1 (en) * 2014-10-24 2016-04-28 Abbvie Inc. Methods of Reducing Methylglyoxal (MGO) Modification of Recombinant Proteins in Cell Culture
IL276165B (en) * 2014-12-01 2022-11-01 Amgen Inc Process for manipulating the level of glycan content of a glycoprotein
KR102007930B1 (en) * 2014-12-31 2019-08-06 주식회사 엘지화학 A method for controlling glycosylation of recombinant glycoprotein
WO2016118707A1 (en) 2015-01-21 2016-07-28 Oncobiologics, Inc. Modulation of charge variants in a monoclonal antibody composition
US20160280767A1 (en) * 2015-03-23 2016-09-29 Lonza Ltd. Methods for controlling protein glycosylation
HU231463B1 (en) * 2015-08-04 2024-01-28 Richter Gedeon Nyrt. Method for increasing the galactose content of recombinant proteins
US11401509B2 (en) 2015-11-09 2022-08-02 Bristol-Myers Squibb Company Methods of minimizing glycoprotein agregation in CHO cell production
EP3411401A1 (en) 2016-02-03 2018-12-12 Oncobiologics, Inc. Buffer formulations for enhanced antibody stability
US10519479B1 (en) * 2016-03-15 2019-12-31 Ares Trading S.A. Methods for modifying glycosylation using manganese
US20200347126A1 (en) 2016-08-03 2020-11-05 Fyb 202 Project Gmbh Production of Biosimilar Ustekinumab In CHO Cells
WO2020033827A1 (en) * 2018-08-10 2020-02-13 Genentech, Inc. Cell culture strategies for modulating protein glycosylation
HUP1800376A2 (en) 2018-11-07 2020-05-28 Richter Gedeon Nyrt Method for modifying the glycosylation profile of a recombinant glycoprotein produced in cell culture
MA54248A (en) * 2018-11-13 2021-09-22 Janssen Biotech Inc REGULATION OF TRACE METALS DURING ANTI-CD38 ANTIBODY PRODUCTION
CN111321188A (en) * 2018-12-17 2020-06-23 嘉和生物药业有限公司 Formula for modifying antibody glycoform, cell culture method and application in industrial production
WO2020190031A1 (en) 2019-03-18 2020-09-24 (주)지플러스 생명과학 Transgenic plant with suppressed expression of cgl1 and cgl2 and method for producing target protein using same
KR102348638B1 (en) 2019-11-21 2022-01-11 (주)지플러스 생명과학 Antibody produced by using afucosylated n.benthamiana and uses thereof
CN112779307B (en) * 2021-01-11 2022-04-19 苏州药明生物技术有限公司 Method for two-stage regulation of CHO expression exogenous protein glycoform
WO2022154762A1 (en) * 2021-01-18 2022-07-21 Turgut İlaçlari A.Ş. Method of producing adalimumab
WO2023021532A1 (en) * 2021-08-20 2023-02-23 Dr. Reddy’S Laboratories Limited A process to produce a pharmaceutical composition
WO2023244746A1 (en) 2022-06-15 2023-12-21 Abbvie Inc. Risankizumab compositions

Family Cites Families (523)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
USRE30985E (en) 1978-01-01 1982-06-29 Serum-free cell culture media
US5179017A (en) 1980-02-25 1993-01-12 The Trustees Of Columbia University In The City Of New York Processes for inserting DNA into eucaryotic cells and for producing proteinaceous materials
US4399216A (en) 1980-02-25 1983-08-16 The Trustees Of Columbia University Processes for inserting DNA into eucaryotic cells and for producing proteinaceous materials
US4634665A (en) 1980-02-25 1987-01-06 The Trustees Of Columbia University In The City Of New York Processes for inserting DNA into eucaryotic cells and for producing proteinaceous materials
EP0101681B1 (en) 1981-09-08 1994-12-14 The Rockefeller University Antibody to a mediator activity composition and its use in a pharmaceutical composition
US4510245A (en) 1982-11-18 1985-04-09 Chiron Corporation Adenovirus promoter system
US4560655A (en) 1982-12-16 1985-12-24 Immunex Corporation Serum-free cell culture medium and process for making same
US4657866A (en) 1982-12-21 1987-04-14 Sudhir Kumar Serum-free, synthetic, completely chemically defined tissue culture media
GB8308235D0 (en) 1983-03-25 1983-05-05 Celltech Ltd Polypeptides
DD266710A3 (en) 1983-06-06 1989-04-12 Ve Forschungszentrum Biotechnologie Process for the biotechnical production of alkaline phosphatase
US4767704A (en) 1983-10-07 1988-08-30 Columbia University In The City Of New York Protein-free culture medium
US4704366A (en) 1984-06-22 1987-11-03 Bio-Rad Laboratories, Inc. Process for binding IgG to protein A
US5672347A (en) 1984-07-05 1997-09-30 Genentech, Inc. Tumor necrosis factor antagonists and their use
DE3431140A1 (en) 1984-08-24 1986-03-06 Behringwerke Ag, 3550 Marburg ENHANCER FOR EUKARYOTIC EXPRESSION SYSTEMS
US4879231A (en) 1984-10-30 1989-11-07 Phillips Petroleum Company Transformation of yeasts of the genus pichia
IL73883A (en) 1984-12-20 1990-12-23 Yeda Res & Dev Monoclonal antibodies against tnf-alpha,hybridomas producing them and method for the purification of tnf-alpha
US5168062A (en) 1985-01-30 1992-12-01 University Of Iowa Research Foundation Transfer vectors and microorganisms containing human cytomegalovirus immediate-early promoter-regulatory DNA sequence
GB8516415D0 (en) 1985-06-28 1985-07-31 Celltech Ltd Culture of animal cells
US5672502A (en) 1985-06-28 1997-09-30 Celltech Therapeutics Limited Animal cell culture
EP0212489B1 (en) 1985-08-16 1994-11-30 The Rockefeller University Anabolic activity modulator and uses thereof
IT206190Z2 (en) 1985-12-17 1987-07-13 Iveco Fiat IMPROVING THE BODYWORK OF A HEAVY VEHICLE
US4968615A (en) 1985-12-18 1990-11-06 Ciba-Geigy Corporation Deoxyribonucleic acid segment from a virus
US4925796A (en) 1986-03-07 1990-05-15 Massachusetts Institute Of Technology Method for enhancing glycoprotein stability
US5681718A (en) 1986-03-14 1997-10-28 Celltech Limited Methods for enhanced production of tissue plasminogen activator in cell culture using alkanoic acids or salts thereof
US4927762A (en) 1986-04-01 1990-05-22 Cell Enterprises, Inc. Cell culture medium with antioxidant
GB8610600D0 (en) 1986-04-30 1986-06-04 Novo Industri As Transformation of trichoderma
DE3631229A1 (en) 1986-09-13 1988-03-24 Basf Ag MONOCLONAL ANTIBODIES AGAINST HUMAN TUMORNESCROSE FACTOR (TNF) AND THEIR USE
US4801687A (en) 1986-10-27 1989-01-31 Bioprobe International, Inc. Monoclonal antibody purification process using protein A
US5045468A (en) 1986-12-12 1991-09-03 Cell Enterprises, Inc. Protein-free culture medium which promotes hybridoma growth
US4877608A (en) 1987-11-09 1989-10-31 Rorer Pharmaceutical Corporation Pharmaceutical plasma protein formulations in low ionic strength media
US6238891B1 (en) 1987-11-18 2001-05-29 Cetus Oncology Corporation Method of increasing product expression through solute stress
US5118796A (en) 1987-12-09 1992-06-02 Centocor, Incorporated Efficient large-scale purification of immunoglobulins and derivatives
NZ229922A (en) 1988-07-18 1992-04-28 Chiron Corp Monoclonal antibodies specifically binding cachectin (tumor necrosis factor) and compositions
US6048728A (en) 1988-09-23 2000-04-11 Chiron Corporation Cell culture medium for enhanced cell growth, culture longevity, and product expression
EP0435911B1 (en) 1988-09-23 1996-03-13 Cetus Oncology Corporation Cell culture medium for enhanced cell growth, culture longevity and product expression
US5126250A (en) 1988-09-28 1992-06-30 Eli Lilly And Company Method for the reduction of heterogeneity of monoclonal antibodies
GB8823869D0 (en) 1988-10-12 1988-11-16 Medical Res Council Production of antibodies
EP0366043B1 (en) 1988-10-24 1994-03-30 Otsuka Pharmaceutical Co., Ltd. Monoclonal antibody
JP2919890B2 (en) 1988-11-11 1999-07-19 メディカル リサーチ カウンスル Single domain ligand, receptor consisting of the ligand, method for producing the same, and use of the ligand and the receptor
EP0374510B1 (en) 1988-12-19 1997-01-15 American Cyanamid Company Products for the treatment of endotoxic shock in a mammal
US5047335A (en) 1988-12-21 1991-09-10 The Regents Of The University Of Calif. Process for controlling intracellular glycosylation of proteins
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
DE4009603A1 (en) 1989-03-30 1990-10-04 Leybold Ag Lock chamber for substrate
US4933435A (en) 1989-04-05 1990-06-12 Bioprobe International Antibody purification process
US5169936A (en) 1989-04-14 1992-12-08 Biogen, Inc. Protein purification on immobilized metal affinity resins effected by elution using a weak ligand
EP0402226A1 (en) 1989-06-06 1990-12-12 Institut National De La Recherche Agronomique Transformation vectors for yeast yarrowia
US6448380B2 (en) 1989-08-07 2002-09-10 Peptech Limited Tumor necrosis factor antibodies
US5959087A (en) 1989-08-07 1999-09-28 Peptide Technology, Ltd. Tumour necrosis factor binding ligands
AU640400B2 (en) 1989-08-07 1993-08-26 Peptide Technology Ltd. Tumour necrosis factor binding ligands
GB8921123D0 (en) 1989-09-19 1989-11-08 Millar Ann B Treatment of ards
US5859205A (en) 1989-12-21 1999-01-12 Celltech Limited Humanised antibodies
GB8928874D0 (en) 1989-12-21 1990-02-28 Celltech Ltd Humanised antibodies
US6075181A (en) 1990-01-12 2000-06-13 Abgenix, Inc. Human antibodies derived from immunized xenomice
US6150584A (en) 1990-01-12 2000-11-21 Abgenix, Inc. Human antibodies derived from immunized xenomice
US6673986B1 (en) 1990-01-12 2004-01-06 Abgenix, Inc. Generation of xenogeneic antibodies
US5945098A (en) 1990-02-01 1999-08-31 Baxter International Inc. Stable intravenously-administrable immune globulin preparation
DE4037604A1 (en) 1990-04-25 1991-10-31 Bayer Ag Use of anti-TNF antibodies to treat ischaemia and its sequelae - esp. to increase survival rate after myocardial infarct and transplants
US5378612A (en) 1990-05-11 1995-01-03 Juridical Foundation The Chemo-Sero-Therapeutic Research Institute Culture medium for production of recombinant protein
US5110913A (en) 1990-05-25 1992-05-05 Miles Inc. Antibody purification method
US5096816A (en) 1990-06-05 1992-03-17 Cetus Corporation In vitro management of ammonia's effect on glycosylation of cell products through pH control
GB9014932D0 (en) 1990-07-05 1990-08-22 Celltech Ltd Recombinant dna product and method
GB9015198D0 (en) 1990-07-10 1990-08-29 Brien Caroline J O Binding substance
DK0814159T3 (en) 1990-08-29 2005-10-24 Genpharm Int Transgenic, non-human animals capable of forming heterologous antibodies
US5770429A (en) 1990-08-29 1998-06-23 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5633425A (en) 1990-08-29 1997-05-27 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5625126A (en) 1990-08-29 1997-04-29 Genpharm International, Inc. Transgenic non-human animals for producing heterologous antibodies
US6255458B1 (en) 1990-08-29 2001-07-03 Genpharm International High affinity human antibodies and human antibodies against digoxin
US5545806A (en) 1990-08-29 1996-08-13 Genpharm International, Inc. Ransgenic non-human animals for producing heterologous antibodies
US5789650A (en) 1990-08-29 1998-08-04 Genpharm International, Inc. Transgenic non-human animals for producing heterologous antibodies
US7084260B1 (en) 1996-10-10 2006-08-01 Genpharm International, Inc. High affinity human antibodies and human antibodies against human antigens
US5661016A (en) 1990-08-29 1997-08-26 Genpharm International Inc. Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
US6300129B1 (en) 1990-08-29 2001-10-09 Genpharm International Transgenic non-human animals for producing heterologous antibodies
US5122469A (en) 1990-10-03 1992-06-16 Genentech, Inc. Method for culturing Chinese hamster ovary cells to improve production of recombinant proteins
GB9022545D0 (en) 1990-10-17 1990-11-28 Wellcome Found Culture medium
GB9022543D0 (en) 1990-10-17 1990-11-28 Wellcome Found Antibody production
GB9022547D0 (en) 1990-10-17 1990-11-28 Wellcome Found Purified immunoglobulin
GB9109645D0 (en) 1991-05-03 1991-06-26 Celltech Ltd Recombinant antibodies
GB2279077B (en) 1990-12-21 1995-06-14 Celltech Ltd Therapeutic compositions comprising recombinant antibodies specific for the TNFalpha
US5994510A (en) 1990-12-21 1999-11-30 Celltech Therapeutics Limited Recombinant antibodies specific for TNFα
GB9028123D0 (en) 1990-12-28 1991-02-13 Erba Carlo Spa Monoclonal antibodies against human tumor necrosis factor alpha
EP0575545B1 (en) 1991-03-15 2003-05-21 Amgen Inc. Pegylation of polypeptides
US6277969B1 (en) 1991-03-18 2001-08-21 New York University Anti-TNF antibodies and peptides of human tumor necrosis factor
US5698195A (en) 1991-03-18 1997-12-16 New York University Medical Center Methods of treating rheumatoid arthritis using chimeric anti-TNF antibodies
US20060246073A1 (en) 1991-03-18 2006-11-02 Knight David M Anti-TNF antibodies and peptides of human tumor necrosis factor
US5656272A (en) 1991-03-18 1997-08-12 New York University Medical Center Methods of treating TNF-α-mediated Crohn's disease using chimeric anti-TNF antibodies
US7192584B2 (en) 1991-03-18 2007-03-20 Centocor, Inc. Methods of treating psoriasis with anti-TNF antibodies
US20070298040A1 (en) 1991-03-18 2007-12-27 Centocor, Inc. Methods of treating seronegative arthropathy with anti-TNF antibodies
US20040120952A1 (en) 2000-08-07 2004-06-24 Centocor, Inc Anti-TNF antibodies and peptides of human tumor necrosis factor
EP1857554A1 (en) 1991-03-18 2007-11-21 New York University Monoclonal and chimeric antibodies specific for human tumor necrosis factor
DK0604418T3 (en) 1991-03-29 1999-06-14 Immunex Corp Isolated viral protein cytokine antagonists
US5328985A (en) 1991-07-12 1994-07-12 The Regents Of The University Of California Recombinant streptavidin-protein chimeras useful for conjugation of molecules in the immune system
IE922287A1 (en) 1991-07-15 1993-01-27 Wellcome Found Production of antibodies
DK0605522T3 (en) 1991-09-23 2000-01-17 Medical Res Council Process for producing humanized antibodies
GB9122820D0 (en) 1991-10-28 1991-12-11 Wellcome Found Stabilised antibodies
WO1994026910A1 (en) 1993-05-12 1994-11-24 Xoma Corporation Immunotoxins comprising gelonin and an antibody
WO1993011793A1 (en) 1991-12-17 1993-06-24 Schering Corporation Use of the combination of anti-tumor necrosis factor plus interleukin-6 to treat septic shock
US5605923A (en) 1992-04-02 1997-02-25 Smithkline Beecham Corporation Compounds useful for treating inflammatory diseases and inhibiting production of tumor necrosis factor
ES2301158T3 (en) 1992-07-24 2008-06-16 Amgen Fremont Inc. XENOGENIC ANTIBODY PRODUCTION.
DE69321909T2 (en) 1992-08-28 1999-04-01 Bayer Ag Use of anti-TNF monoclonal antibodies for the treatment of bacterial meningitis
US6270766B1 (en) 1992-10-08 2001-08-07 The Kennedy Institute Of Rheumatology Anti-TNF antibodies and methotrexate in the treatment of arthritis and crohn's disease
DE69312077T2 (en) 1992-10-08 1997-10-30 Kennedy Inst Of Rheumatology TREATMENT OF AUTOIMMUNE AND INFLAMMATION DISEASES
AU682156B2 (en) 1992-10-15 1997-09-25 Dana-Farber Cancer Institute, Inc. Treatment of insulin resistance in obesity linked type II diabetes using antagonists to TNF-alpha function
JPH06292592A (en) 1993-02-09 1994-10-21 Snow Brand Milk Prod Co Ltd Production of glycoprotein
ES2159529T5 (en) 1993-03-05 2011-03-09 Bayer Corporation ANTI-TNF ALFA HUMAN MONOCLONAL ANTIBODIES.
DE4307508A1 (en) 1993-03-10 1994-09-15 Knoll Ag Use of anti-TNF antibodies as a medicine in the treatment of heart failure (heart muscle weakness)
EP0754225A4 (en) 1993-04-26 2001-01-31 Genpharm Int Transgenic non-human animals capable of producing heterologous antibodies
DE69405102T2 (en) 1993-06-03 1998-01-15 Therapeutic Antibodies Inc ANTIBODY FRAGMENTS IN THERAPY
WO1995011317A1 (en) 1993-10-19 1995-04-27 The Scripps Research Institute Synthetic human neutralizing monoclonal antibodies to human immunodeficiency virus
EP0659766A1 (en) 1993-11-23 1995-06-28 Schering-Plough Human monoclonal antibodies against human cytokines and methods of making and using such antibodies
NZ278607A (en) 1994-02-07 1999-05-28 Knoll Ag Use of tnf antagonists for treating disorders involving elevated serum levels of il-6 wherein the serum levels are 500pg/ml or above
EP0666312A1 (en) 1994-02-08 1995-08-09 Wolfgang A. Renner Process for the improvement of mammalian cell growth
US5429746A (en) 1994-02-22 1995-07-04 Smith Kline Beecham Corporation Antibody purification
EP0748338A4 (en) 1994-03-04 2001-03-28 Merck & Co Inc In vitro antibody affinity maturation using alanine scanning mutagenesis
US5856179A (en) 1994-03-10 1999-01-05 Genentech, Inc. Polypeptide production in animal cell culture
US5945311A (en) 1994-06-03 1999-08-31 GSF--Forschungszentrumfur Umweltund Gesundheit Method for producing heterologous bi-specific antibodies
WO1996000081A1 (en) 1994-06-24 1996-01-04 Immunex Corporation Controlled release polypeptide compositions and methods of treating inflammatory bowel disease
ZA955642B (en) 1994-07-07 1997-05-06 Ortho Pharma Corp Lyophilized imaging agent formulation
US5561053A (en) 1994-08-05 1996-10-01 Genentech, Inc. Method for selecting high-expressing host cells
SE503424C2 (en) 1994-11-14 1996-06-10 Pharmacia Ab Process for purification of recombinant coagulation factor VIII
US5641870A (en) 1995-04-20 1997-06-24 Genentech, Inc. Low pH hydrophobic interaction chromatography for antibody purification
EP0822830B1 (en) 1995-04-27 2008-04-02 Amgen Fremont Inc. Human anti-IL-8 antibodies, derived from immunized xenomice
AU2466895A (en) 1995-04-28 1996-11-18 Abgenix, Inc. Human antibodies derived from immunized xenomice
US6656466B1 (en) 1995-06-06 2003-12-02 Genetech, Inc. Human tumor necrosis factor—immunoglobulin(TNFR1-IgG1) chimera composition
US5721121A (en) 1995-06-06 1998-02-24 Genentech, Inc. Mammalian cell culture process for producing a tumor necrosis factor receptor immunoglobulin chimeric protein
US5705364A (en) 1995-06-06 1998-01-06 Genentech, Inc. Mammalian cell culture process
US6113898A (en) 1995-06-07 2000-09-05 Idec Pharmaceuticals Corporation Human B7.1-specific primatized antibodies and transfectomas expressing said antibodies
WO1997004801A1 (en) 1995-07-27 1997-02-13 Genentech, Inc. Stabile isotonic lyophilized protein formulation
JP4306813B2 (en) 1995-09-19 2009-08-05 アスビオファーマ株式会社 New method for culturing animal cells
AU6873396A (en) 1995-10-16 1997-05-07 Unilever N.V. A bifunctional or bivalent antibody fragment analogue
US6090382A (en) 1996-02-09 2000-07-18 Basf Aktiengesellschaft Human antibodies that bind human TNFα
AR005035A1 (en) 1995-12-11 1999-04-07 Merck Patent Ges Mit Beschränkter Haftung PROCEDURE TO PREPARE RECOMBINANT PROTEINS IN E. COLI, BY FERMENTATION WITH GREAT CONCENTRATION OF CELLS.
WO1997029131A1 (en) 1996-02-09 1997-08-14 Basf Aktiengesellschaft HUMAN ANTIBODIES THAT BIND HUMAN TNF$g(a)
ATE222598T1 (en) 1996-04-19 2002-09-15 Nestle Sa HUMAN IMMORTALIZED COLON EPITHELIAL CELLS
TW491855B (en) 1996-08-07 2002-06-21 Csl Ltd Purification of immunoglobulins
JP2000517188A (en) 1996-08-30 2000-12-26 ライフ テクノロジーズ,インコーポレイテッド Serum-free mammalian cell culture medium and uses thereof
US20040171152A1 (en) 1996-10-10 2004-09-02 Invitrogen Corporation Animal cell culture media comprising non-animal or plant-derived nutrients
DE69739673D1 (en) 1996-11-27 2009-12-31 Genentech Inc Affinity Purification of Polypeptide Proteins on a Protein A Matrix
CA2722378C (en) 1996-12-03 2015-02-03 Amgen Fremont Inc. Human antibodies that bind tnf.alpha.
GB9625175D0 (en) 1996-12-04 1997-01-22 Medi Cult As Serum-free cell culture media
US5804420A (en) 1997-04-18 1998-09-08 Bayer Corporation Preparation of recombinant Factor VIII in a protein free medium
PE84799A1 (en) 1997-04-28 1999-09-16 Lilly Co Eli IMPROVED METHODS FOR THE PROCESSING OF ACTIVATED C PROTEIN
US6235883B1 (en) 1997-05-05 2001-05-22 Abgenix, Inc. Human monoclonal antibodies to epidermal growth factor receptor
US6171586B1 (en) 1997-06-13 2001-01-09 Genentech, Inc. Antibody formulation
DK0999853T3 (en) 1997-06-13 2003-04-22 Genentech Inc Stabilized antibody formulation
US6475725B1 (en) 1997-06-20 2002-11-05 Baxter Aktiengesellschaft Recombinant cell clones having increased stability and methods of making and using the same
EP0994903B1 (en) 1997-06-24 2005-05-25 Genentech, Inc. Methods and compositions for galactosylated glycoproteins
US20040191256A1 (en) 1997-06-24 2004-09-30 Genentech, Inc. Methods and compositions for galactosylated glycoproteins
US20020045207A1 (en) 1997-10-31 2002-04-18 Lynne A. Krummen Glycoprotein production process
US20040136986A1 (en) 1997-10-31 2004-07-15 Genentech, Inc. Methods and compositions comprising glycoprotein glycoforms
EP1028751B1 (en) 1997-10-31 2008-12-31 Genentech, Inc. Methods and compositions comprising glycoprotein glycoforms
ES2252876T5 (en) 1997-12-03 2014-04-03 Roche Diagnostics Gmbh Process to prepare polypeptides with adequate glycolization
WO1999032605A1 (en) 1997-12-19 1999-07-01 Novo Nordisk A/S Method for producing heterologous proteins in eukaryotic cells on an industrial scale using nucleotide-manipulating agents
EP2261229A3 (en) 1998-04-20 2011-03-23 GlycArt Biotechnology AG Glycosylation engineering of antibodies for improving antibody-dependent cellular cytotoxicity
WO1999057246A1 (en) 1998-05-01 1999-11-11 Life Technologies, Inc. Animal cell culture media comprising non-animal or plant-derived nutrients
EP1308455B9 (en) 1998-05-06 2006-06-14 Genentech, Inc. A composition comprising anti-HER2 antibodies
IL139035A0 (en) 1998-05-06 2001-11-25 Genentech Inc Protein purification by ion exchange chromatography
DE19821933C1 (en) 1998-05-15 1999-11-11 Disetronic Licensing Ag Device for administering an injectable product
DE19822031C2 (en) 1998-05-15 2000-03-23 Disetronic Licensing Ag Auto injection device
US6528286B1 (en) 1998-05-29 2003-03-04 Genentech, Inc. Mammalian cell culture process for producing glycoproteins
US6406909B1 (en) 1998-07-10 2002-06-18 Chugai Seiyaku Kabushiki Kaisha Serum-free medium for culturing animal cells
US7883704B2 (en) 1999-03-25 2011-02-08 Abbott Gmbh & Co. Kg Methods for inhibiting the activity of the P40 subunit of human IL-12
US6914128B1 (en) 1999-03-25 2005-07-05 Abbott Gmbh & Co. Kg Human antibodies that bind human IL-12 and methods for producing
AU3633000A (en) 1999-03-26 2000-10-16 Human Genome Sciences, Inc. Neutrokine-alpha binding proteins and methods based thereon
EP2270148A3 (en) 1999-04-09 2011-06-08 Kyowa Hakko Kirin Co., Ltd. Method for controlling the activity of immunologically functional molecule
US7297680B2 (en) 1999-04-15 2007-11-20 Crucell Holland B.V. Compositions of erythropoietin isoforms comprising Lewis-X structures and high sialic acid content
JP4543131B2 (en) 1999-04-26 2010-09-15 ジェネンテック, インコーポレイテッド Cell culture methods for glycoproteins
AU4314900A (en) 1999-04-28 2000-11-17 Yamanouchi Pharmaceutical Co., Ltd. Parenteral medicinal composition containing humanized monoclonal antibody fragment and method for stabilizing the same
AT409379B (en) 1999-06-02 2002-07-25 Baxter Ag MEDIUM FOR PROTEIN- AND SERUM-FREE CELL CULTURE
CA2379413A1 (en) 1999-07-30 2001-02-08 Genentech, Inc. Charged filtration membranes and uses therefor
ATE313634T1 (en) 1999-09-27 2006-01-15 Genentech Inc METHOD FOR PRODUCING RECOMBINANT PROTEINS USING APOPTOSIS INHIBITORS
AR026743A1 (en) 1999-12-09 2003-02-26 Pharmacia Ab PRODUCTION OF PEPTIDES
KR20010056451A (en) 1999-12-15 2001-07-04 윤재승 Arginine-enriched medium used for mass-producing recombinant protein in animal cell culture
JP4516711B2 (en) 1999-12-28 2010-08-04 中外製薬株式会社 Stable antibody composition and injection preparation
ATE329018T1 (en) 2000-02-08 2006-06-15 Genentech Inc IMPROVED GALACTOSYLATION OF RECOMBINANT PROTEINS
EP2338515A3 (en) 2000-02-10 2011-11-16 Abbott Laboratories Antibodies that bind human interleukin-18 and methods of making and using
GB0003231D0 (en) 2000-02-11 2000-04-05 Medi Cult As Cell culture media
JP2001218840A (en) 2000-02-14 2001-08-14 Kanegafuchi Chem Ind Co Ltd Adsorbent for transforming growth factor. beta, adsorbing/removing method, and adsorber
WO2001077362A1 (en) 2000-04-06 2001-10-18 Chugai Seiyaku Kabushiki Kaisha Immunoassay of anti-hm1.24 antibody
CA2405912A1 (en) 2000-04-12 2001-10-18 Human Genome Sciences, Inc. Albumin fusion proteins
ATE440959T1 (en) 2000-06-28 2009-09-15 Glycofi Inc METHOD FOR PRODUCING MODIFIED GLYCOPROTEINS
US7598055B2 (en) 2000-06-28 2009-10-06 Glycofi, Inc. N-acetylglucosaminyltransferase III expression in lower eukaryotes
US7449308B2 (en) 2000-06-28 2008-11-11 Glycofi, Inc. Combinatorial DNA library for producing modified N-glycans in lower eukaryotes
EP1298217A4 (en) 2000-07-05 2004-09-08 Mitsubishi Pharma Corp Process for producing glycoprotein
WO2002011753A1 (en) 2000-08-04 2002-02-14 Chugai Seiyaku Kabushiki Kaisha Protein injection preparations
US20060018907A1 (en) 2000-08-07 2006-01-26 Centocor, Inc. Anti-TNF antibodies and peptides of human tumor necrosis factor
US7288390B2 (en) 2000-08-07 2007-10-30 Centocor, Inc. Anti-dual integrin antibodies, compositions, methods and uses
UA81743C2 (en) 2000-08-07 2008-02-11 Центокор, Инк. HUMAN MONOCLONAL ANTIBODY WHICH SPECIFICALLY BINDS TUMOR NECROSIS FACTOR ALFA (TNFα), PHARMACEUTICAL MIXTURE CONTAINING THEREOF, AND METHOD FOR TREATING ARTHRITIS
US20050249735A1 (en) 2000-08-07 2005-11-10 Centocor, Inc. Methods of treating ankylosing spondylitis using anti-TNF antibodies and peptides of human tumor necrosis factor
US6635448B2 (en) 2000-08-21 2003-10-21 Clonexdevelopment, Inc. Methods and compositions for increasing protein yield from a cell culture
BR0114373A (en) 2000-10-02 2004-02-17 Novo Nordisk As Eukaryotic host cell, methods for the production of fvii and fviia or variants thereof, a vitamin ke-dependent protein of a fvii-producing eukaryotic host cell or analog, recombinant vector, fvii and fviia or variants thereof, and , recombinant vitamin K dependent protein
US20090151023A1 (en) 2000-11-13 2009-06-11 Viktor Kuvshinov Transformation system for Camelina sativa
DE60230858D1 (en) 2001-02-15 2009-03-05 Centocor Inc CHEMICALLY DEFINED MEDIUM FOR CULTIVATED MAMMALIAN CELLS
US20140186361A1 (en) 2012-09-07 2014-07-03 Coherus Biosciences, Inc. Stable Aqueous Formulations of Adalimumab
US20030096414A1 (en) 2001-03-27 2003-05-22 Invitrogen Corporation Culture medium for cell growth and transfection
AU2002256005A1 (en) 2001-03-27 2002-10-08 Smithkline Beecham Corporation Control of glycoforms in igg
US7189820B2 (en) 2001-05-24 2007-03-13 Human Genome Sciences, Inc. Antibodies against tumor necrosis factor delta (APRIL)
BR0206160A (en) 2001-05-25 2004-10-26 Abbott Gmbh & Co Kg Use of anti-TNF antibodies as medicines in the treatment of septic disorders of anemic patients.
CA2385745C (en) 2001-06-08 2015-02-17 Abbott Laboratories (Bermuda) Ltd. Methods of administering anti-tnf.alpha. antibodies
EP1404813A4 (en) 2001-06-13 2004-11-24 Genentech Inc Methods of culturing animal cells and polypeptide production in animal cells
ES2674888T3 (en) 2001-06-26 2018-07-04 Amgen Inc. OPGL antibodies
CN1555411A (en) 2001-08-03 2004-12-15 ���迨�����\���ɷݹ�˾ Antibody glycosylation variants having increased antibody-dependent cellular cytotoxicity
GB0119520D0 (en) 2001-08-10 2001-10-03 Owen Mumford Ltd Improvements relating to injection devices
CN1309825C (en) 2001-10-02 2007-04-11 诺和诺德医疗保健公司 Method for production of recombinant proteins in eukaryote cells
US7125843B2 (en) 2001-10-19 2006-10-24 Neose Technologies, Inc. Glycoconjugates including more than one peptide
HUP0600342A3 (en) 2001-10-25 2011-03-28 Genentech Inc Glycoprotein compositions
AU2002340787B2 (en) 2001-11-12 2009-09-17 Monash University Peptide purification by means of metal ion affinity chromatography
PL370550A1 (en) 2001-11-28 2005-05-30 Sandoz Ag Method for producing a recombinant polypeptide
CN1596302A (en) 2001-11-28 2005-03-16 桑多斯有限公司 Cell culture process
WO2003046162A2 (en) 2001-11-28 2003-06-05 Polymun Scientific Immunbiologische Forschung Gmbh Process for the production of polypeptides in mammalian cell cultures
US7473680B2 (en) 2001-11-28 2009-01-06 Neose Technologies, Inc. Remodeling and glycoconjugation of peptides
AU2002359816B2 (en) 2001-12-21 2006-07-13 Immunex Corporation Methods for purifying protein
JP4460302B2 (en) 2002-02-05 2010-05-12 ジェネンテック インコーポレイテッド Protein purification method
US20030161828A1 (en) 2002-02-19 2003-08-28 Abbott Gmbh & Co. Kg Use of TNF antagonists as drugs for the treatment of patients with an inflammatory reaction and without suffering from total organ failure
CA2417689C (en) 2002-03-05 2006-05-09 F. Hoffmann-La Roche Ag Improved methods for growing mammalian cells in vitro
ES2557741T3 (en) 2002-03-27 2016-01-28 Immunex Corporation Procedures to increase polypeptide production
US20030190710A1 (en) 2002-03-28 2003-10-09 Devries Ruth L. Control of glycoforms in IgG
JPWO2003085118A1 (en) 2002-04-09 2005-08-11 協和醗酵工業株式会社 Method for producing antibody composition
US20040009172A1 (en) 2002-04-26 2004-01-15 Steven Fischkoff Use of anti-TNFalpha antibodies and another drug
WO2003102132A2 (en) 2002-04-26 2003-12-11 Genetech, Inc. Non-affinity purification of proteins
US20030206898A1 (en) 2002-04-26 2003-11-06 Steven Fischkoff Use of anti-TNFalpha antibodies and another drug
US20040029229A1 (en) 2002-05-20 2004-02-12 Reeves Philip J. High level protein expression system
AU2003249055B2 (en) 2002-07-15 2008-06-26 Immunex Corporation Methods and media for controlling sialylation of proteins produced by mammalian cells
US20090280065A1 (en) 2006-04-10 2009-11-12 Willian Mary K Uses and Compositions for Treatment of Psoriasis
PL213925B1 (en) 2002-07-19 2013-05-31 Abbott Biotech Ltd Treatment of tnf ó related disorders
US20040033228A1 (en) 2002-08-16 2004-02-19 Hans-Juergen Krause Formulation of human antibodies for treating TNF-alpha associated disorders
US6924124B1 (en) 2002-08-23 2005-08-02 Immunex Corporation Feeding strategies for cell culture
US6974681B1 (en) 2002-08-23 2005-12-13 Immunex Corporation Cell culture performance with vanadate
US7067279B1 (en) 2002-08-23 2006-06-27 Immunex Corporation Cell culture performance with betaine
EP1578774B1 (en) 2002-09-18 2013-11-06 Danisco US Inc. Purification of immunoglobulins
US6890736B1 (en) 2002-09-20 2005-05-10 Immunex Corporation Methods for producing proteins in cultured cells
MY150740A (en) 2002-10-24 2014-02-28 Abbvie Biotechnology Ltd Low dose methods for treating disorders in which tnf? activity is detrimental
JP4500683B2 (en) 2002-11-01 2010-07-14 バイエル・ヘルスケア・エルエルシー Macromolecule concentration method
US20040086532A1 (en) 2002-11-05 2004-05-06 Allergan, Inc., Botulinum toxin formulations for oral administration
US20040162414A1 (en) 2002-11-22 2004-08-19 Santora Ling C. Method for reducing or preventing modification of a polypeptide in solution
US20040101939A1 (en) 2002-11-22 2004-05-27 Santora Ling C. Method for reducing or preventing modification of a polypeptide in solution
WO2004055164A2 (en) 2002-12-13 2004-07-01 Abgenix, Inc. System and method for stabilizing antibodies with histidine
CN101857851A (en) 2002-12-23 2010-10-13 布里斯托尔-迈尔斯斯奎布公司 Mammalian cell culture processes for protein production
AU2003303394B2 (en) * 2002-12-23 2009-02-19 Bristol-Myers Squibb Company Product quality enhancement in mammalian cell culture processes for protein production
PT1601697E (en) 2003-02-28 2007-09-04 Lonza Biologics Plc Antibody purification by protein a and ion exchange chromatography
DE602004008451T2 (en) 2003-05-01 2008-05-15 Dsm Ip Assets B.V. PROCESS FOR THE PREPARATION OF BIOLOGICAL SUBSTANCES BY PERFUSION CULTURING OF SUSPENDED ANIMAL CELLS
SI1623019T1 (en) 2003-05-15 2010-10-29 Wyeth Llc Restricted glucose feed for animal cell culture
GB2404665B (en) 2003-08-08 2005-07-06 Cambridge Antibody Tech Cell culture
EP1673452B1 (en) 2003-10-10 2015-12-23 Novo Nordisk Health Care AG Method for large-scale production of a polypeptide in eukaryote cells
FR2861080B1 (en) 2003-10-20 2006-02-17 Lab Francais Du Fractionnement ANTIBODIES HAVING AN OPTIMIZED FUCOSE AND GALACTOSE RATE
WO2005042569A1 (en) 2003-10-24 2005-05-12 Amgen, Inc. Process for purifying proteins in a hydrophobic interaction chromatography flow-through fraction
US20050100965A1 (en) 2003-11-12 2005-05-12 Tariq Ghayur IL-18 binding proteins
DE10355251A1 (en) 2003-11-26 2005-06-23 Merck Patent Gmbh Water-based pharmaceutical preparation for treatment of tumors has active ingredient effective against receptor of endothelial growth factor receptor
WO2005056600A2 (en) 2003-12-08 2005-06-23 The Government Of The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Monoclonal antibodies that bind or neutralize dengue virus
PL1703893T3 (en) 2003-12-23 2012-09-28 Genentech Inc Novel anti-il 13 antibodies and uses thereof
EP1697414A2 (en) 2003-12-23 2006-09-06 Applied Research Systems ARS Holding N.V. Process for the production of tumor necrosis factor-binding proteins
US20080058507A1 (en) 2004-02-11 2008-03-06 Hui Liu Method For The Removal Of Aggregate Proteins From Recombinant Samples Using Ion Exchange Chromatography
AU2005211890B2 (en) 2004-02-12 2011-07-28 Merck Patent Gmbh Highly concentrated liquid formulations of anti-EGFR antibodies
EP1718386A1 (en) 2004-02-27 2006-11-08 GE Healthcare Bio-Sciences AB A process for the purification of antibodies
CN1234725C (en) 2004-04-07 2006-01-04 陈志南 High performance quick purifying method for preparing piecewise antibody
TWI439284B (en) 2004-04-09 2014-06-01 Abbvie Biotechnology Ltd Multiple-variable dose regimen for treating tnfα-related disorders
WO2005111627A2 (en) 2004-04-15 2005-11-24 Massachusetts Institute Of Technology Methods and products related to the improved analysis of carbohydrates
AU2005233387B2 (en) 2004-04-15 2011-05-26 Glycofi, Inc. Production of galactosylated glycoproteins in lower eukaryotes
US20060127950A1 (en) 2004-04-15 2006-06-15 Massachusetts Institute Of Technology Methods and products related to the improved analysis of carbohydrates
RU2006138181A (en) 2004-05-04 2008-06-10 Ново Нордиск Хелс Кеа Аг (Ch) O-RELATED Glycoforms of Polypeptides and a Method for Their Production
AU2005269763A1 (en) 2004-07-21 2006-02-09 Glycofi, Inc. Immunoglobulins comprising predominantly a Gal2GlcNac2Man3GlcNac2 glycoform
US20060223147A1 (en) 2004-08-05 2006-10-05 Kyowa Hakko Kogyo Co., Ltd., Process for producing glycoprotein composition
TWI384069B (en) 2004-08-27 2013-02-01 Pfizer Ireland Pharmaceuticals Production of polypeptides
ES2570006T3 (en) 2004-09-30 2016-05-13 Bayer Healthcare Llc Devices and methods of continuous integrated preparation of biological molecules
WO2006041970A2 (en) 2004-10-08 2006-04-20 Abbott Biotechnology Ltd. Treatment of respiratory syncytial virus (rsv) infection
US20060194301A1 (en) 2004-10-09 2006-08-31 Doctor Bhupendra P Large-scale production of human serum butyrylcholinesterase as a bioscavenger
KR101243601B1 (en) 2004-10-21 2013-03-20 지이 헬스케어 바이오-사이언시스 에이비 Chromatography ligand
US7029982B1 (en) 2004-10-21 2006-04-18 Sharp Laboratories Of America, Inc. Method of affecting RRAM characteristics by doping PCMO thin films
US20060094104A1 (en) 2004-10-29 2006-05-04 Leopold Grillberger Animal protein-free media for cultivation of cells
US20080160577A1 (en) 2005-02-04 2008-07-03 Glaxo Group Limited Optimization of Heterologous Polypeptide Expression
US8530192B2 (en) 2005-02-11 2013-09-10 Novo Nordisk Healthcare Ag Production of a polypeptide in a serum-free cell culture liquid containing plant protein hydrolysate
WO2006099308A2 (en) 2005-03-11 2006-09-21 Wyeth A method of weak partitioning chromatography
US20060252672A1 (en) 2005-04-05 2006-11-09 Betenbaugh Michael J Protein N-glycosylation of eukaryotic cells using dolichol-linked oligosaccharide synthesis pathway, other N-gylosylation-increasing methods, and engineered hosts expressing products with increased N-glycosylation
CA2604877A1 (en) 2005-04-11 2006-10-19 J.P. Sercel Associates Inc. Protein purification
US9963510B2 (en) 2005-04-15 2018-05-08 Macrogenics, Inc. Covalent diabodies and uses thereof
WO2006113743A2 (en) 2005-04-18 2006-10-26 Massachusetts Institute Of Technology Compositions and methods for rna interference with sialidase expression and uses thereof
CA2606270A1 (en) 2005-04-19 2006-10-26 Massachusetts Institute Of Technology Amphiphilic polymers and methods of use thereof
CA2903138A1 (en) 2005-05-16 2006-11-23 Abbvie Biotechnology Ltd. Use of tnfa inhibitor for treatment of erosive polyarthritis
US20060275867A1 (en) 2005-06-03 2006-12-07 Veronique Chotteau Process
EP1888638A2 (en) 2005-06-03 2008-02-20 Genentech, Inc. Method of producing antibodies with modified fucosylation level
ES2543685T3 (en) 2005-06-30 2015-08-21 Janssen Biotech, Inc. Methods and compositions with improved therapeutic activity
CA2615532C (en) 2005-07-26 2016-06-28 Sangamo Biosciences, Inc. Targeted integration and expression of exogenous nucleic acid sequences
US7612181B2 (en) 2005-08-19 2009-11-03 Abbott Laboratories Dual variable domain immunoglobulin and uses thereof
US20070041905A1 (en) 2005-08-19 2007-02-22 Hoffman Rebecca S Method of treating depression using a TNF-alpha antibody
CA2619825A1 (en) 2005-08-19 2007-03-01 Centocor, Inc. Proteolysis resistant antibody preparations
RS57549B1 (en) 2005-08-26 2018-10-31 Ares Trading Sa Process for the preparation of glycosylated interferon beta
CA2620515A1 (en) 2005-09-02 2007-03-08 Glycofi, Inc. Immunoglobulins comprising predominantly a glcnacman3glcnac2 glycoform
PE20070796A1 (en) 2005-10-24 2007-08-15 Wyeth Corp PROTEIN PRODUCTION METHOD USING ANTI-SENESCENCE COMPOUNDS
KR20140012160A (en) 2005-11-01 2014-01-29 애브비 바이오테크놀로지 리미티드 Methods and compositions for diagnosing ankylosing spondylitis using biomarkers
US20080206246A1 (en) 2006-04-05 2008-08-28 Ravetch Jeffrey V Polypeptides with enhanced anti-inflammatory and decreased cytotoxic properties and relating methods
US8470318B2 (en) 2005-11-07 2013-06-25 The Rockefeller University Polypeptides with enhanced anti-inflammatory and decreased cytotoxic properties and relating methods
EP2455100A3 (en) 2005-11-07 2012-11-07 The Rockefeller University Reagents, methods and systems for selecting a cytotoxic antibody or variant thereof
CA2630811C (en) 2005-12-08 2016-05-31 Amgen Inc. Improved production of glycoproteins using manganese
US20070212770A1 (en) 2006-01-04 2007-09-13 Baxter International Inc. Oligopeptide-free cell culture media
US20070190057A1 (en) 2006-01-23 2007-08-16 Jian Wu Methods for modulating mannose content of recombinant proteins
US20070202051A1 (en) 2006-02-10 2007-08-30 Pari Gmbh Aerosols for sinunasal drug delivery
KR20090005315A (en) 2006-04-05 2009-01-13 애보트 바이오테크놀로지 리미티드 Antibody purification
US10167332B2 (en) 2006-04-05 2019-01-01 The Rockefeller University Polypeptides with enhanced anti-inflammatory and decreased cytotoxic properties and relating methods
EP2666472A3 (en) 2006-04-10 2014-04-02 Abbott Biotechnology Ltd Uses and compositions for treatment of psoriatic arthritis
US20080118496A1 (en) 2006-04-10 2008-05-22 Medich John R Uses and compositions for treatment of juvenile rheumatoid arthritis
US9605064B2 (en) 2006-04-10 2017-03-28 Abbvie Biotechnology Ltd Methods and compositions for treatment of skin disorders
US20090317399A1 (en) 2006-04-10 2009-12-24 Pollack Paul F Uses and compositions for treatment of CROHN'S disease
EP2012586A4 (en) 2006-04-10 2010-08-18 Abbott Biotech Ltd Uses and compositions for treatment of ankylosing spondylitis
US9399061B2 (en) 2006-04-10 2016-07-26 Abbvie Biotechnology Ltd Methods for determining efficacy of TNF-α inhibitors for treatment of rheumatoid arthritis
US7846724B2 (en) 2006-04-11 2010-12-07 Hoffmann-La Roche Inc. Method for selecting CHO cell for production of glycosylated antibodies
US20080131374A1 (en) 2006-04-19 2008-06-05 Medich John R Uses and compositions for treatment of rheumatoid arthritis
CA2652578A1 (en) 2006-05-19 2007-11-29 Glycofi, Inc. Erythropoietin compositions
US20080311043A1 (en) 2006-06-08 2008-12-18 Hoffman Rebecca S Uses and compositions for treatment of psoriatic arthritis
US20100021451A1 (en) 2006-06-08 2010-01-28 Wong Robert L Uses and compositions for treatment of ankylosing spondylitis
EP2069387A4 (en) 2006-06-14 2011-02-02 Glaxosmithkline Llc Methods for purifying antibodies using ceramic hydroxyapatite
CN103316403B (en) 2006-06-30 2017-05-24 艾伯维生物技术有限公司 Automatic injection device
WO2008008360A1 (en) 2006-07-13 2008-01-17 Wyeth Production of glycoproteins
WO2008028974A1 (en) 2006-09-08 2008-03-13 Novo Nordisk A/S Methods of optimizing chromatographic separation of polypeptides
PL2073842T3 (en) 2006-09-10 2015-06-30 Glycotope Gmbh Use of human cells of myeloid leukaemia origin for expression of antibodies
TW201516149A (en) 2006-09-13 2015-05-01 Abbvie Inc Cell culture improvements
US8911964B2 (en) 2006-09-13 2014-12-16 Abbvie Inc. Fed-batch method of making human anti-TNF-alpha antibody
EP2500416A1 (en) 2006-09-13 2012-09-19 Abbott Laboratories Cell culture improvements
WO2008036600A2 (en) 2006-09-18 2008-03-27 Genentech, Inc. Methods of protein production
CL2007002880A1 (en) 2006-10-06 2008-05-09 Amgen Inc STABLE FORMULATION THAT INCLUDES AN ACETIC ACID STAMP, A GLUTAMIC ACID STAMP OR A SUCCINIC ACID STAMP WITH A PH OF 4.5 TO 7, AT LEAST A EXCIPIENT THAT INCLUDES A SUGAR OR A POLYOL, AND AN ANTI-RECEPTOR ANTIBODY OF FACTOR D
WO2008057240A2 (en) 2006-10-27 2008-05-15 Abbott Biotechnology Ltd. Crystalline anti-htnfalpha antibodies
EP2091969A4 (en) 2006-10-27 2010-05-12 Univ Rockefeller Polypeptides with enhanced anti-inflammatory and decreased cytotoxic properties and relating methods
ES2541546T3 (en) 2006-11-03 2015-07-21 Wyeth Llc Substances that inhibit glycolysis in cell culture
WO2008068879A1 (en) 2006-12-06 2008-06-12 Jcr Pharmaceuticals Co., Ltd. Method for production of human erythropoietin
US20080226635A1 (en) 2006-12-22 2008-09-18 Hans Koll Antibodies against insulin-like growth factor I receptor and uses thereof
CN101563460A (en) 2006-12-22 2009-10-21 弗·哈夫曼-拉罗切有限公司 Selection method
RU2466189C2 (en) * 2006-12-28 2012-11-10 Сентокор Орто Байотек Инк. POLYPEPTIDE EXPRESSION VECTOR WITH SYALIDASE ACTIVITY, METHOD FOR PROVIDING SYALIDASE ACTIVITY IN CELL CULTURE AND METHOD FOR CONTROLLING PROPERTIES OF Fc-CONTAINING MOLECULES EXPRESSED IN CELL LINE
US7691980B2 (en) 2007-01-09 2010-04-06 Bio-Rad Laboratories, Inc. Enhanced capacity and purification of antibodies by mixed mode chromatography in the presence of aqueous-soluble nonionic organic polymers
WO2008087184A2 (en) 2007-01-17 2008-07-24 Merck Serono S.A. Process for the purification of fc-containing proteins
EP2115126B1 (en) 2007-03-02 2015-04-08 Wyeth LLC Use of copper and glutamate in cell culture for production of polypeptides
RU2476442C2 (en) 2007-03-29 2013-02-27 Эббот Лэборетриз Crystalline human il-12 antibodies
US20100209434A1 (en) 2007-03-30 2010-08-19 Medimmune, Llc Antibody formulation
CN101680014B (en) 2007-04-03 2018-09-28 奥克西雷恩英国有限公司 The glycosylation of molecule
WO2008130924A2 (en) 2007-04-16 2008-10-30 Momenta Pharmaceuticals, Inc. Isotopically-labeled glycans
WO2008128225A1 (en) 2007-04-16 2008-10-23 Momenta Pharmaceuticals, Inc. Multi-dimensional chromatographic methods for separating n-glycans
US9182467B2 (en) 2007-04-16 2015-11-10 Momenta Pharmaceuticals, Inc. Comparative analysis of protein conformations by using 2D NOESY NMR spectra
EP2135093B1 (en) 2007-04-16 2015-04-15 Momenta Pharmaceuticals, Inc. Analysis of phosphorylated glycans, glcopeptides or glycoproteins by imac
EP2511705A1 (en) 2007-04-16 2012-10-17 Momenta Pharmaceuticals, Inc. Methods related to cell surface glycosylation
AU2008243007A1 (en) 2007-04-16 2008-10-30 Momenta Pharmaceuticals, Inc. Characterization of N-glycans using exoglycosidases
US20100113294A1 (en) 2007-04-16 2010-05-06 Momenta Pharmaceuticals, Inc. Defined glycoprotein products and related methods
WO2008128218A1 (en) 2007-04-16 2008-10-23 Momenta Pharmaceuticals, Inc. Characterization of n-glycan mixtures by nuclear magnetic resonance
ES2367210T3 (en) 2007-04-16 2011-10-31 Momenta Pharmaceuticals, Inc. MS METHODS TO EVALUATE GLICANS.
WO2008128216A1 (en) 2007-04-16 2008-10-23 Momenta Pharmaceuticals, Inc. Methods for labeling glycans
WO2008128220A1 (en) 2007-04-16 2008-10-23 Momenta Pharmaceuticals, Inc. Proteolytic release of glycans
WO2008128230A1 (en) 2007-04-16 2008-10-23 Momenta Pharmaceuticals, Inc. Reference glycoprotein products and related methods
TW200902708A (en) 2007-04-23 2009-01-16 Wyeth Corp Methods of protein production using anti-senescence compounds
WO2008135498A2 (en) 2007-05-04 2008-11-13 Novo Nordisk A/S Prevention of protein degradation in mammalian cell cultures
EP1988101A1 (en) 2007-05-04 2008-11-05 Novo Nordisk A/S Improvement of factor VIII polypeptide titers in cell cultures
AU2008251405B2 (en) 2007-05-11 2012-05-17 Amgen Inc. Improved feed media
WO2008150491A2 (en) 2007-05-31 2008-12-11 Abbott Laboratories BIOMARKERS PREDICTIVE OF THE RESPONSIVENESS TO TNFα INHIBITORS IN AUTOIMMUNE DISORDERS
WO2008150490A2 (en) 2007-06-01 2008-12-11 Abbott Biotechnology Ltd. Uses and compositions for treatment of psoriasis and crohn's disease
US8999337B2 (en) 2007-06-11 2015-04-07 Abbvie Biotechnology Ltd. Methods for treating juvenile idiopathic arthritis by inhibition of TNFα
WO2008154014A2 (en) 2007-06-11 2008-12-18 Amgen Inc. A method for culturing mammalian cells to improve recombinant protein production
DK3597659T3 (en) 2007-07-09 2023-04-03 Genentech Inc Prevention of disulfide bond reduction during recombinant production of polypeptides
CA2693771A1 (en) 2007-07-13 2009-01-22 Abbott Biotechnology Ltd. Methods and compositions for pulmonary administration of a tnf.alpha. inhibitor
KR100897159B1 (en) 2007-07-26 2009-05-14 보령제약 주식회사 A plant recombinant human CTLA4Ig and a method for producing the same
JP2010535771A (en) 2007-08-08 2010-11-25 アボット・ラボラトリーズ Compositions and methods for crystallizing antibodies
ES2657055T3 (en) 2007-08-09 2018-03-01 Wyeth Llc Use of perfusion to improve the production of a batch-fed cell culture in bioreactors
MX2010002269A (en) 2007-08-28 2010-03-25 Abbott Biotech Ltd Compositions and methods comprising binding proteins for adalimumab.
EP2031064A1 (en) 2007-08-29 2009-03-04 Boehringer Ingelheim Pharma GmbH & Co. KG Method for increasing protein titres
CN101784670A (en) 2007-08-31 2010-07-21 弗·哈夫曼-拉罗切有限公司 Glycosylation profile analysis
US8273723B2 (en) 2007-10-12 2012-09-25 Sigma-Aldrich Co. Llc Compositions and methods for improved glycoprotein sialylation
EP2050764A1 (en) 2007-10-15 2009-04-22 sanofi-aventis Novel polyvalent bispecific antibody format and uses thereof
WO2009058307A1 (en) 2007-10-29 2009-05-07 University Of Georgia Research Foundation, Inc. In vivo isotopic labeling method for quantitative glycomics
WO2009058769A1 (en) 2007-10-30 2009-05-07 Schering Corporation Purification of antibodies containing hydrophobic variants
US8883146B2 (en) 2007-11-30 2014-11-11 Abbvie Inc. Protein formulations and methods of making same
US20130195888A1 (en) 2007-11-30 2013-08-01 Abbvie Ultrafiltration and diafiltration formulation methods for protein processing
KR20190045414A (en) 2007-11-30 2019-05-02 애브비 바이오테크놀로지 리미티드 Protein formulations and methods of making same
SG10201503304RA (en) 2007-12-27 2015-06-29 Baxter Int Cell culture processes
CA2710779C (en) 2007-12-31 2017-06-20 Bayer Schering Pharma Aktiengesellschaft Antibodies to tnf.alpha.
CA2710333A1 (en) 2008-01-03 2009-07-09 Abbott Biotechnology Ltd. Predicting long-term efficacy of a compound in the treatment of psoriasis
DK2240595T4 (en) 2008-01-03 2019-10-07 Cornell Res Foundation Inc Glycosylated protein expression in prokaryotes
RU2502800C2 (en) 2008-01-15 2013-12-27 Эббви Инк. Improved expression vectors of mammals and their use
CN104188911A (en) 2008-01-15 2014-12-10 Abbvie德国有限责任两合公司 Powdered protein compositions and methods of making same
MX2010008364A (en) 2008-01-30 2010-08-23 Abbott Lab Compositions and methods for crystallizing antibody fragments.
TWI472339B (en) 2008-01-30 2015-02-11 Genentech Inc Composition comprising antibody that binds to domain ii of her2 and acidic variants thereof
AU2009222115A1 (en) 2008-02-29 2009-09-11 Biogen Idec Ma Inc. Purified immunoglobulin fusion proteins and methods of their purification
WO2009114641A1 (en) 2008-03-11 2009-09-17 Genentech, Inc. Antibodies with enhanced adcc function
WO2009118662A2 (en) 2008-03-24 2009-10-01 Abbott Biotechnology Ltd. Methods and compositions for treating bone loss
JP2011517410A (en) 2008-04-08 2011-06-09 アミリス バイオテクノロジーズ, インコーポレイテッド Heterologous sequence expression
CA2723197C (en) 2008-05-02 2017-09-19 Seattle Genetics, Inc. Methods and compositions for making antibodies and antibody derivatives with reduced core fucosylation
WO2009135656A1 (en) 2008-05-06 2009-11-12 Lonza Biologics Plc. A method for the purification of antibodies using displacement chromatography
JP2011523853A (en) 2008-06-03 2011-08-25 アボット・ラボラトリーズ Dual variable domain immunoglobulins and uses thereof
WO2010016943A2 (en) 2008-08-08 2010-02-11 Biogen Idec Ma Inc. Nutrient monitoring and feedback control for increased bioproduct production
ES2527943T5 (en) 2008-08-14 2019-03-07 Genentech Inc Methods to remove a contaminant using protein displacement ion exchange membrane chromatography
US20100069617A1 (en) 2008-09-12 2010-03-18 Ge Healthcare Bio-Sciences Ab Enhanced protein aggregate removal by mixed mode chromatography on hydrophobic interaction media in the presence of protein-excluded zwitterions
CN102216452B (en) 2008-09-26 2013-08-21 尤里卡治疗公司 Cell lines and proteins with variant glycosylation pattern
WO2010043703A1 (en) 2008-10-17 2010-04-22 Dsm Ip Assets B.V. Removal of host cell proteins
CN102257005A (en) 2008-10-20 2011-11-23 雅培制药有限公司 Viral inactivation during purification of antibodies
CN105111309A (en) 2008-10-20 2015-12-02 Abbvie公司 Isolation and purification of antibodies using protein an appinity chromatography
US20100150864A1 (en) 2008-10-20 2010-06-17 Abbott Laboratories, Inc. Antibodies that bind to il-18 and methods of purifying the same
GB0821100D0 (en) 2008-11-18 2008-12-24 Hansa Medical Ab Antibodies
US9103821B2 (en) 2008-12-19 2015-08-11 Momenta Pharmaceuticals, Inc. Methods related to modified glycans
US8729241B2 (en) 2008-12-19 2014-05-20 Momenta Pharmaceuticals, Inc. Characterization of O-linked glycans
US8614297B2 (en) 2008-12-22 2013-12-24 Hoffmann-La Roche Inc. Anti-idiotype antibody against an antibody against the amyloid β peptide
WO2010080062A1 (en) 2009-01-08 2010-07-15 Ge Healthcare Bio-Sciences Ab Separation method using single polymer phase systems
KR20110119702A (en) 2009-01-22 2011-11-02 모멘타 파머슈티컬스 인코포레이티드 Galactose-alpha-1,3-galactose-containing n-glycans in glycoprotein products derived from cho cells
EP2400981A4 (en) 2009-02-26 2013-02-27 Lpath Inc Humanized platelet activating factor antibody design using anti-lipid antibody templates
WO2010102114A1 (en) 2009-03-05 2010-09-10 Biogen Idec Ma Inc. Purification of immunoglobulins
EP2233499A1 (en) 2009-03-26 2010-09-29 CSL Behring AG Antibody composition with altered Fab sialylation
JP4981192B2 (en) 2009-04-01 2012-07-18 株式会社イーベック Monoclonal antibody that binds to a specific discontinuous epitope present in the AD1 region of human cytomegalovirus gB glycoprotein and antigen-binding fragment thereof
EP2421892A1 (en) 2009-04-20 2012-02-29 Pfizer Inc. Control of protein glycosylation and compositions and methods relating thereto
KR101721906B1 (en) 2009-04-29 2017-03-31 애브비 바이오테크놀로지 리미티드 Automatic injection device
WO2010127069A1 (en) 2009-04-29 2010-11-04 Schering Corporation Antibody purification
JP2012526121A (en) 2009-05-04 2012-10-25 アボツト・バイオテクノロジー・リミテツド Stable high protein concentration formulation of human anti-TNF alpha antibody
AU2010254215A1 (en) 2009-05-26 2011-12-01 Momenta Pharmaceuticals, Inc. Production of glycoproteins
EP2435556B1 (en) 2009-05-28 2016-05-25 Boehringer Ingelheim International GmbH Method for a rational cell culturing process
WO2010136209A1 (en) 2009-05-28 2010-12-02 ETH Zürich N-glycan core beta-galactosyltransferase and uses thereof
AU2010256455A1 (en) 2009-06-05 2012-01-19 Momenta Pharmaceuticals, Inc. Methods of modulating fucosylation of glycoproteins
EP2451936B1 (en) 2009-07-06 2019-08-28 F.Hoffmann-La Roche Ag Method of culturing eukaryotic cells
BRPI1010119A2 (en) 2009-07-24 2016-03-15 Hoffmann La Roche "antibody composition production method and use of analytical ion exchange chromatography"
US8945895B2 (en) 2009-07-31 2015-02-03 Baxter International Inc. Methods of purifying recombinant ADAMTS13 and other proteins and compositions thereof
WO2011015926A1 (en) 2009-08-03 2011-02-10 Avesthagen Limited A process of fermentation, purification and production of recombinant soluble tumour necrosis factor alfa receptor (tnfr) - human igg fc fusion protein
WO2011019620A1 (en) 2009-08-10 2011-02-17 Genentech, Inc. Antibodies with enhanced adcc function
ES2793348T3 (en) 2009-08-11 2020-11-13 Hoffmann La Roche Protein production in glutamine-free cell culture media
WO2011019622A1 (en) 2009-08-14 2011-02-17 Genentech, Inc. Cell culture methods to make antibodies with enhanced adcc function
WO2011024025A1 (en) 2009-08-28 2011-03-03 Avesthagen Limited An erythropoietin analogue and a method thereof
US9540426B2 (en) 2009-10-06 2017-01-10 Bristol-Myers Squibb Company Mammalian cell culture processes for protein production
US8470552B2 (en) 2009-10-12 2013-06-25 Keck Graduate Institute Strategy to reduce lactic acid production and control PH in animal cell culture
EP2325296A1 (en) 2009-11-20 2011-05-25 LEK Pharmaceuticals d.d. Production of glycoproteins with low N-glycolylneuraminic acid (Neu5Gc) content
US9096879B2 (en) 2009-11-24 2015-08-04 Biogen Ma Inc. Method of supplementing culture media to prevent undesirable amino acid substitutions
US10087236B2 (en) 2009-12-02 2018-10-02 Academia Sinica Methods for modifying human antibodies by glycan engineering
WO2011069056A2 (en) 2009-12-04 2011-06-09 Momenta Pharmaceuticals, Inc. Antennary fucosylation in glycoproteins from cho cells
CN104193798A (en) 2009-12-18 2014-12-10 Csl有限公司 Method of purifying polypeptides
JP2013518590A (en) 2010-02-02 2013-05-23 アボツト・バイオテクノロジー・リミテツド Methods and compositions for predicting responsiveness to treatment with a TNF-α inhibitor
WO2011095596A1 (en) 2010-02-04 2011-08-11 Vivalis Fed-batch process using concentrated cell culture medium for the efficient production of biologics in eb66 cells
EP2534169A1 (en) 2010-02-12 2012-12-19 DSM IP Assets B.V. Single unit antibody purification
WO2011110598A1 (en) 2010-03-10 2011-09-15 F. Hoffmann-La Roche Ag Method for purifying immunoglobulin solutions
WO2011112892A1 (en) 2010-03-12 2011-09-15 Purecircle Usa Inc. High-purity steviol glycosides
MX2012011648A (en) 2010-04-07 2012-11-29 Momenta Pharmaceuticals Inc High mannose glycans.
MX346731B (en) 2010-04-23 2017-03-30 Genentech Inc * Production of heteromultimeric proteins.
US20110262965A1 (en) 2010-04-23 2011-10-27 Life Technologies Corporation Cell culture medium comprising small peptides
CA2795461C (en) 2010-04-26 2018-04-24 Novartis Ag Improved cell cultivation process
MX2012012526A (en) 2010-04-26 2012-11-23 Novartis Ag Improved cell culture medium.
DK2576580T3 (en) 2010-05-28 2016-10-17 Hoffmann La Roche LOWERING THE lactate AND INCREASE polypeptide production by downregulation of the expression of lactate dehydrogenase, and PYRUVATDEHYDROGENASEKINASE
DK2575884T3 (en) 2010-06-03 2018-09-03 Abbvie Biotechnology Ltd APPLICATIONS AND COMPOSITIONS TO TREAT HIDRADENITIS SUPPURATIVA (HS)
CA2808116C (en) 2010-07-30 2016-06-07 Pfizer Inc. Tandem purification of proteins
CN103080300B (en) 2010-08-05 2015-11-25 安姆根有限公司 Increase the productive rate of cell culture and the dipeptides of vigor
DE102010038990A1 (en) 2010-08-06 2012-02-09 Robert Bosch Gmbh Shaft bearing device for a hand tool
WO2012030512A1 (en) 2010-09-03 2012-03-08 Percivia Llc. Flow-through protein purification process
CN103119448A (en) 2010-09-17 2013-05-22 Abbvie公司 Raman spectroscopy for bioprocess operations
SG188616A1 (en) 2010-09-20 2013-04-30 Abbvie Inc Purification of antibodies using simulated moving bed chromatography
US20120258496A1 (en) 2010-09-27 2012-10-11 Boehringer Ingelheim International Gmbh Production of low fucose antibodies in h4-ii-e rat cells
EA201390461A1 (en) 2010-10-08 2013-09-30 Кадила Хелзкэр Лимитед VECTOR OF EXPRESSION FOR HIGHLY EFFICIENT EXPRESSION OF RECOMBINANT PROTEINS
JP6023715B2 (en) 2010-10-11 2016-11-09 アッヴィ・バハマズ・リミテッド Protein purification method
JP5918697B2 (en) 2010-10-15 2016-05-18 Jcrファーマ株式会社 Method for producing a glycoprotein in which the non-reducing end of the sugar chain is a mannose residue
EP2450375A1 (en) 2010-11-09 2012-05-09 Sandoz Gmbh Cell culture medium and process for protein expression, said medium and process comprising a PAM inhibitor
RU2015154965A (en) 2010-11-11 2019-01-16 Эббви Байотекнолоджи Лтд. IMPROVED HIGH-CONCENTRATED LIQUID PRODUCTS ANTIBODIES AGAINST TNF ALPHA
EP2640483A1 (en) 2010-11-15 2013-09-25 Biogen Idec Inc. Enrichment and concentration of select product isoforms by overloaded bind and elute chromatography
US20120149878A1 (en) 2010-12-08 2012-06-14 Gillespie Ronald Protein purification
BR112013017417A2 (en) 2011-01-07 2016-09-27 Abbvie Inc anti-il-12 / il-23 antibodies and uses thereof
TW201309330A (en) 2011-01-28 2013-03-01 Abbott Lab Compositions containing glycosylated antibodies and uses thereof
AR085302A1 (en) 2011-02-24 2013-09-18 Sanofi Sa METHOD OF PRODUCTION OF STIRATED ANTIBODIES
CN103597073B (en) 2011-03-06 2019-06-07 默克雪兰诺有限公司 Low fucose cell line and its application
US20120230913A1 (en) 2011-03-10 2012-09-13 Board Of Regents, The University Of Texas System Protein nanoparticle dispersions
WO2012125735A1 (en) 2011-03-15 2012-09-20 Abott Laboratories An integrated approach to the isolation and purification of antibodies
TWI671315B (en) 2011-03-28 2019-09-11 法商賽諾菲公司 Dual variable region antibody-like binding proteins having cross-over binding region orientation
EP2511293A1 (en) 2011-04-13 2012-10-17 LEK Pharmaceuticals d.d. A method for controlling the main complex N-glycan structures and the acidic variants and variability in bioprocesses producing recombinant proteins
US9133493B2 (en) 2011-04-21 2015-09-15 Amgen Inc. Method for culturing mammalian cells to improve recombinant protein production
EP2702077A2 (en) 2011-04-27 2014-03-05 AbbVie Inc. Methods for controlling the galactosylation profile of recombinantly-expressed proteins
ES2560470T3 (en) 2011-04-29 2016-02-19 Biocon Research Limited A method to reduce the heterogeneity of antibodies and a production process of said antibodies
US20140108084A1 (en) 2012-10-12 2014-04-17 Crestron Electronics, Inc. Initiating Schedule Management Via Radio Frequency Beacons
CN103517707A (en) 2011-04-29 2014-01-15 西莱克塔生物科技公司 Controlled release of immunosuppressants from synthetic nanocarriers
RU2592680C2 (en) 2011-04-29 2016-07-27 Биокон Рисерч Лимитед Method for reduction of accumulation of lactate during culturing and method of producing antibody
JP6236383B2 (en) 2011-05-13 2017-11-22 バイオジェン・エムエイ・インコーポレイテッドBiogen MA Inc. Methods for preventing and removing trisulfide bonds
HUE033279T2 (en) 2011-07-01 2017-11-28 Amgen Inc Mammalian cell culture
WO2013006461A1 (en) 2011-07-01 2013-01-10 Biogen Idec Ma Inc. Cholesterol-based media supplementals for cell culture
WO2013004841A1 (en) 2011-07-06 2013-01-10 Genmab A/S Modulation of complement-dependent cytotoxicity through modifications of the c-terminus of antibody heavy chains
US9475858B2 (en) 2011-07-08 2016-10-25 Momenta Pharmaceuticals, Inc. Cell culture process
GB201112429D0 (en) 2011-07-19 2011-08-31 Glaxo Group Ltd Antigen-binding proteins with increased FcRn binding
WO2013013013A2 (en) 2011-07-21 2013-01-24 Alnylam Pharmaceuticals, Inc. Compositions and methods for producing modified glycoproteins
KR20140101331A (en) 2011-08-10 2014-08-19 라보라토이레 프란카이즈 듀 프락티온네먼트 에트 데스 바이오테크놀로지스 Highly galactosylated antibodies
WO2013095738A2 (en) 2011-09-27 2013-06-27 Mapp Biopharmaceutical, Inc. Monoclonal antibodies with altered affinities for human fcyri, fcyrllla, and c1q proteins
US20140288278A1 (en) 2011-10-31 2014-09-25 Joseph Nti-Gyabaah Chromatography process for resolving heterogeneous antibody aggregates
RU2014121884A (en) 2011-11-02 2015-12-10 Дженентек, Инк. CHROMATOGRAPHY OF OVERLOAD AND ELOURATION
JP6322872B2 (en) 2011-12-19 2018-05-16 ザ ロックフェラー ユニバーシティー Non-sialylated anti-inflammatory peptide
SG11201405475UA (en) 2012-03-07 2014-10-30 Cadila Healthcare Ltd Pharmaceutical formulations of tnf-alpha antibodies
SG10201701224UA (en) 2012-03-12 2017-04-27 Merck Patent Gmbh Removal of protein aggregates from biopharmaceutical preparations in a flowthrough mode
US9067990B2 (en) 2013-03-14 2015-06-30 Abbvie, Inc. Protein purification using displacement chromatography
WO2013158273A1 (en) 2012-04-20 2013-10-24 Abbvie Inc. Methods to modulate c-terminal lysine variant distribution
WO2013158275A1 (en) 2012-04-20 2013-10-24 Abbvie Inc. Cell culture methods to reduce acidic species
WO2013158279A1 (en) 2012-04-20 2013-10-24 Abbvie Inc. Protein purification methods to reduce acidic species
US20140154270A1 (en) 2012-05-21 2014-06-05 Chen Wang Purification of non-human antibodies using kosmotropic salt enhanced protein a affinity chromatography
US20130336957A1 (en) 2012-05-21 2013-12-19 Abbvie, Inc. Novel purification of human, humanized, or chimeric antibodies using protein a affinity chromatography
WO2013176754A1 (en) 2012-05-24 2013-11-28 Abbvie Inc. Novel purification of antibodies using hydrophobic interaction chromatography
EP2855745A4 (en) 2012-06-01 2016-01-20 Momenta Pharmaceuticals Inc Methods related to adalimumab
WO2013186230A1 (en) 2012-06-12 2013-12-19 Boehringer Ingelheim International Gmbh Pharmaceutical formulation for a therapeutic antibody
US20150210753A1 (en) 2012-07-26 2015-07-30 Momenta Pharmaceuticals, Inc. Glycoproteins with anti-inflammatory properties
EP2885639B1 (en) 2012-08-17 2019-04-10 MorphoSys AG Complex-specific antibodies and antibody fragments and their use
US9512214B2 (en) 2012-09-02 2016-12-06 Abbvie, Inc. Methods to control protein heterogeneity
AU2013309506A1 (en) 2012-09-02 2015-03-12 Abbvie Inc. Methods to control protein heterogeneity
WO2014052360A2 (en) 2012-09-26 2014-04-03 Momenta Pharmaceuticals, Inc. Glycoprotein preparations
EP2931749B1 (en) 2012-12-17 2019-04-24 Laboratoire Francais du Fractionnement et des Biotechnologies Societe Anonyme Use of monoclonal antibodies for the treatment of inflammation and bacterial infections
WO2014099636A1 (en) 2012-12-18 2014-06-26 Merck Sharp & Dohme Corp. Liquid formulations for an anti-tnf alpha antibody
EP2956480B1 (en) 2013-02-13 2019-09-04 Laboratoire Français du Fractionnement et des Biotechnologies Highly galactosylated anti-tnf-alpha antibodies and uses thereof
KR20150125994A (en) 2013-03-08 2015-11-10 뉴클론 바이오로직스 피티와이 엘티디 A cell expression system
WO2014151878A2 (en) 2013-03-14 2014-09-25 Abbvie Inc. Methods for modulating protein glycosylation profiles of recombinant protein therapeutics using monosaccharides and oligosacharides
US8956830B2 (en) 2013-03-14 2015-02-17 Momenta Pharmaceuticals, Inc. Methods of cell culture
US20140271633A1 (en) 2013-03-14 2014-09-18 Abbvie Inc. Mammalian cell culture performance through surfactant supplementation of feed media
US9217168B2 (en) 2013-03-14 2015-12-22 Momenta Pharmaceuticals, Inc. Methods of cell culture
WO2014159579A1 (en) 2013-03-14 2014-10-02 Abbvie Inc. MUTATED ANTI-TNFα ANTIBODIES AND METHODS OF THEIR USE
US9677105B2 (en) 2013-03-14 2017-06-13 Momenta Pharmaceuticals, Inc. Methods of cell culture
US20140271622A1 (en) 2013-03-14 2014-09-18 Momenta Pharmaceuticals, Inc. Methods of cell culture
US9017687B1 (en) 2013-10-18 2015-04-28 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same using displacement chromatography
CA2899308C (en) 2013-03-14 2017-04-18 Abbvie Inc. Low acidic species adalimumab compositions and uses thereof
JP6472786B2 (en) 2013-03-15 2019-02-20 アルダー・バイオファーマシューティカルズ・インコーポレーテッド Antibody purification and purity monitoring
PT2970980T (en) 2013-03-15 2018-11-19 Janssen Biotech Inc Manufacturing methods to control c-terminal lysine, galactose and sialic acid content in recombinant proteins
ES2802274T3 (en) 2013-05-02 2021-01-18 Momenta Pharmaceuticals Inc Sialylated glycoproteins
KR101569783B1 (en) 2013-06-05 2015-11-19 한화케미칼 주식회사 A Method of Antibody Purification
AR096713A1 (en) 2013-06-25 2016-01-27 Cadila Healthcare Ltd PURIFICATION PROCESS FOR MONOCLONAL ANTIBODIES
NZ715246A (en) 2013-07-06 2017-07-28 Cadila Healthcare Ltd Improved process for production of monoclonal antibodies
SI3021833T2 (en) 2013-07-19 2022-08-31 Hexal Aktiengesellschaft Methods and formulations which allow the modulation of immune responses related to the administration of a biopharmaceutical drug
WO2015026846A1 (en) 2013-08-19 2015-02-26 Biogen Idec Ma Inc. Control of protein glycosylation by culture medium supplementation and cell culture process parameters
CA2915948A1 (en) 2013-08-20 2015-02-26 Lek Pharmaceuticals D.D. Cell culture medium and process for controlling a-amidation and/or c-terminal amino acid cleavage of polypeptides
EP3052640A2 (en) 2013-10-04 2016-08-10 AbbVie Inc. Use of metal ions for modulation of protein glycosylation profiles of recombinant proteins
US9085618B2 (en) 2013-10-18 2015-07-21 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same
US8946395B1 (en) 2013-10-18 2015-02-03 Abbvie Inc. Purification of proteins using hydrophobic interaction chromatography
US9181337B2 (en) 2013-10-18 2015-11-10 Abbvie, Inc. Modulated lysine variant species compositions and methods for producing and using the same
WO2015073884A2 (en) 2013-11-15 2015-05-21 Abbvie, Inc. Glycoengineered binding protein compositions
WO2016007764A1 (en) 2014-07-09 2016-01-14 Abbvie Inc. Methods for modulating the glycosylation profile of recombinant proteins using non-commonly used sugars
US20160039924A1 (en) 2014-08-05 2016-02-11 Abbvie Inc. Methods for modulating the glycosylation profile of recombinant proteins using dissolved oxygen
HRP20231408T1 (en) 2014-12-22 2024-02-16 UCB Biopharma SRL Protein manufacture
CN105777895A (en) 2015-03-19 2016-07-20 广东东阳光药业有限公司 Application of sodium phenylbutyrate in purification of antibody acidic peak
CN105777896B (en) 2015-03-19 2019-08-16 广东东阳光药业有限公司 A kind of purification process at antibody acidity peak
CN105777904B (en) 2015-03-23 2019-12-10 广东东阳光药业有限公司 cation exchange chromatography purification method of anti-TNF alpha monoclonal antibody

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
Tebbey et al., MAbs. 2015 Sep 3;7(5):805-11; submiotted by Applicant as Appendix B received 8/4/2015. Paper in case. *

Cited By (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9255143B2 (en) 2011-04-27 2016-02-09 Abbvie Inc. Methods for controlling the galactosylation profile of recombinantly-expressed proteins
US9505834B2 (en) 2011-04-27 2016-11-29 Abbvie Inc. Methods for controlling the galactosylation profile of recombinantly-expressed proteins
US9365645B1 (en) 2011-04-27 2016-06-14 Abbvie, Inc. Methods for controlling the galactosylation profile of recombinantly-expressed proteins
US9150645B2 (en) 2012-04-20 2015-10-06 Abbvie, Inc. Cell culture methods to reduce acidic species
US9708400B2 (en) 2012-04-20 2017-07-18 Abbvie, Inc. Methods to modulate lysine variant distribution
US9334319B2 (en) 2012-04-20 2016-05-10 Abbvie Inc. Low acidic species compositions
US9346879B2 (en) 2012-04-20 2016-05-24 Abbvie Inc. Protein purification methods to reduce acidic species
US9359434B2 (en) 2012-04-20 2016-06-07 Abbvie, Inc. Cell culture methods to reduce acidic species
US9957318B2 (en) 2012-04-20 2018-05-01 Abbvie Inc. Protein purification methods to reduce acidic species
US9683033B2 (en) 2012-04-20 2017-06-20 Abbvie, Inc. Cell culture methods to reduce acidic species
US9193787B2 (en) 2012-04-20 2015-11-24 Abbvie Inc. Human antibodies that bind human TNF-alpha and methods of preparing the same
US9505833B2 (en) 2012-04-20 2016-11-29 Abbvie Inc. Human antibodies that bind human TNF-alpha and methods of preparing the same
US9249182B2 (en) 2012-05-24 2016-02-02 Abbvie, Inc. Purification of antibodies using hydrophobic interaction chromatography
US9234033B2 (en) 2012-09-02 2016-01-12 Abbvie, Inc. Methods to control protein heterogeneity
US9290568B2 (en) 2012-09-02 2016-03-22 Abbvie, Inc. Methods to control protein heterogeneity
US9512214B2 (en) 2012-09-02 2016-12-06 Abbvie, Inc. Methods to control protein heterogeneity
US9499614B2 (en) 2013-03-14 2016-11-22 Abbvie Inc. Methods for modulating protein glycosylation profiles of recombinant protein therapeutics using monosaccharides and oligosaccharides
US9708399B2 (en) 2013-03-14 2017-07-18 Abbvie, Inc. Protein purification using displacement chromatography
US9598667B2 (en) 2013-10-04 2017-03-21 Abbvie Inc. Use of metal ions for modulation of protein glycosylation profiles of recombinant proteins
US9522953B2 (en) 2013-10-18 2016-12-20 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same
US9499616B2 (en) 2013-10-18 2016-11-22 Abbvie Inc. Modulated lysine variant species compositions and methods for producing and using the same
US9688752B2 (en) 2013-10-18 2017-06-27 Abbvie Inc. Low acidic species compositions and methods for producing and using the same using displacement chromatography
US9200069B2 (en) 2013-10-18 2015-12-01 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same
US9200070B2 (en) 2013-10-18 2015-12-01 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same
US9550826B2 (en) 2013-11-15 2017-01-24 Abbvie Inc. Glycoengineered binding protein compositions

Also Published As

Publication number Publication date
US9505834B2 (en) 2016-11-29
US9062106B2 (en) 2015-06-23
US20120276631A1 (en) 2012-11-01
US20160152702A1 (en) 2016-06-02
US9365645B1 (en) 2016-06-14
US20160207992A1 (en) 2016-07-21
US20150050693A1 (en) 2015-02-19
US9090688B2 (en) 2015-07-28
WO2012149197A3 (en) 2013-05-10
TW201708539A (en) 2017-03-01
TWI557228B (en) 2016-11-11
EP2702077A2 (en) 2014-03-05
TW201247871A (en) 2012-12-01
WO2012149197A2 (en) 2012-11-01
US9255143B2 (en) 2016-02-09
US20170051052A1 (en) 2017-02-23

Similar Documents

Publication Publication Date Title
US9505834B2 (en) Methods for controlling the galactosylation profile of recombinantly-expressed proteins
US9206390B2 (en) Methods to control protein heterogeneity
US11459595B2 (en) Methods for increasing mannose content of recombinant proteins
US9388447B2 (en) Method for culturing mammalian cells to improve recombinant protein production
US11946085B2 (en) Methods for increasing mannose content of recombinant proteins
US20160046708A1 (en) Methods to modulate lysine variant distribution
KR20210095723A (en) Flocculation method
WO2006109781A1 (en) Process for production of glycoprotein composition

Legal Events

Date Code Title Description
AS Assignment

Owner name: ABBOTT LABORATORIES, ILLINOIS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:BENGEA, CORNELIA;RIVES, LISA M.;REEL/FRAME:036419/0224

Effective date: 20120621

Owner name: ABBVIE, INC., ILLINOIS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:HOSSLER, PATRICK;REEL/FRAME:036419/0240

Effective date: 20150303

Owner name: ABBVIE INC., ILLINOIS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:ABBOTT LABORATORIES;REEL/FRAME:036463/0598

Effective date: 20120801

STCF Information on status: patent grant

Free format text: PATENTED CASE

CC Certificate of correction
MAFP Maintenance fee payment

Free format text: PAYMENT OF MAINTENANCE FEE, 4TH YEAR, LARGE ENTITY (ORIGINAL EVENT CODE: M1551); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

Year of fee payment: 4

MAFP Maintenance fee payment

Free format text: PAYMENT OF MAINTENANCE FEE, 8TH YEAR, LARGE ENTITY (ORIGINAL EVENT CODE: M1552); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

Year of fee payment: 8